CN114630829A - Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors - Google Patents

Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors Download PDF

Info

Publication number
CN114630829A
CN114630829A CN202080063781.2A CN202080063781A CN114630829A CN 114630829 A CN114630829 A CN 114630829A CN 202080063781 A CN202080063781 A CN 202080063781A CN 114630829 A CN114630829 A CN 114630829A
Authority
CN
China
Prior art keywords
alkyl
radical
cycloalkyl
substituted
independently selected
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202080063781.2A
Other languages
Chinese (zh)
Inventor
张华杰
何成喜
谭锐
容悦
张卫鹏
周祖文
刘洪彬
陈志方
陈岭
杨理君
王云岭
王宪龙
姜立花
林舒
赵兴东
王为波
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chongqing Fushang Yuanchuang Pharmaceutical Technology Co ltd
Original Assignee
Fochon Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fochon Pharmaceuticals Ltd filed Critical Fochon Pharmaceuticals Ltd
Publication of CN114630829A publication Critical patent/CN114630829A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention provides a class of TRK inhibitors, pharmaceutical compositions thereof, and methods of use thereof.

Description

Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
This application claims priority to U.S. provisional application 62/898,817, the entire contents of which are incorporated herein by reference in their entirety.
Technical Field
The present invention relates to a class of compounds and/or pharmaceutically acceptable salts which inhibit protein tyrosine kinases of the TRK family and as medicaments for the treatment of hyperproliferative diseases such as cancer and inflammation, or immune and autoimmune diseases.
Background
Hyperproliferative diseases such as cancer and inflammation have attracted the academic community to provide effective treatments for them. And efforts have been made in this regard to identify and target specific mechanisms that play a role in proliferative diseases.
Tropomyosin Receptor Kinase (TRK), a neurotrophic tyrosine kinase receptor (NTRK), is a transmembrane tyrosine kinase activated by a group of soluble growth factors that may be referred to as neurotrophic factors (NTs). The TRK family includes TRKA, TRKB and TRKC, encoded by the NTRK1, NTRK2 and NTRK3 genes, respectively. The main neurotrophic ligands of the three TRK subtypes are Nerve Growth Factor (NGF) for activating TRKA, brain-derived neurotrophic factor (BDNF) for activating TRKB and neurotrophic factor 3(NT-3) for activating TRKC. Neurotrophins recognize the TRK protein extracellularly, inducing receptor dimerization and phosphorylation, which in turn activates downstream pathways including PI3K, RAS/MAPK/ERK, and PLC- γ.
TRK is expressed primarily in neuronal tissues and regulates neuronal cell survival and differentiation. Deregulation of the TRK pathway includes gene fusion, protein overexpression and single nucleotide changes, which exacerbate many abnormal physiological processes that negatively impact human health. Inhibitors of the NT/TRK signaling pathway have been currently demonstrated to be effective in a variety of preclinical animal models of inflammation and pain. Furthermore, alterations in TRK signaling pathways have been associated with poor prognosis in a variety of malignant solid tumors, such as neuroblastoma, breast cancer, pancreatic cancer, melanoma, multiple myeloma, thyroid cancer, glioblastoma, colorectal cancer, sarcoma, cholangiocarcinoma, non-small cell lung cancer, and the like. Thus, alterations in the NTRK gene can serve as predictive biomarkers for targeted therapy. Ongoing clinical trials have demonstrated that selective TRK inhibitors are beneficial for patients with tumors that have altered NTRK genes.
Therefore, the compounds having TRK inhibitory activity are of great significance for the prevention and treatment of cancer. Although TRK inhibitors have been reported in the literature, such as WO 2006082392, many have short half-lives or are toxic. Accordingly, there remains a need for novel TRK inhibitors that have advantages in at least one of therapeutic efficacy, stability, selectivity, toxicity, and pharmacodynamic characteristics in the treatment of hyperproliferative diseases. Based on the TRK inhibitor, the invention provides a novel TRK inhibitor.
Disclosure of Invention
The invention discloses a novel compound, pharmaceutically acceptable salts and pharmaceutical compositions thereof, and application as a medicament.
In one aspect, the invention provides a compound of formula (I):
Figure BDA0003541883110000021
or a pharmaceutically acceptable salt thereof, wherein:
when X is N and Y is C, as shown in formula (Ia);
Figure BDA0003541883110000022
when X is C, Y is N, as shown in formula (Ib);
Figure BDA0003541883110000023
R1selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with a substituent of (1);
each R2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA1RB1、-ORA1-C(O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(O)ORA1、-OC(O)RA1、-C(O)NRA1RB1、-NRA1C(O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(O)NRA1RB1、-NRA1C(O)ORB1、-NRA1C(O)NRA1RB1、-NRA1C(S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(O)rRA1、-S(O)(=NRE1)RB1、-N=S(O)RA1RB1、-S(O)2ORA1、-OS(O)2RA1、-NRA1S(O)rRB1、-NRA1S(O)(=NRE1)RB1、-S(O)rNRA1RB1、-S(O)(=NRE1)NRA1RB1、-NRA1S(O)2NRA1RB1、-NRA1S(O)(=NRE1)NRA1RB1、-P(O)RA1RB1and-P (O) (OR)A1)(ORB1) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
R3selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
R4selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA2RB2、-ORA2、-C(O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(O)ORA2、-OC(O)RA2、-C(O)NRA2RB2、-NRA2C(O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(O)NRA2RB2、-NRA2C(O)ORB2、-NRA2C(O)NRA2RB2、-NRA2C(S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(O)rRA2、-S(O)(=NRE2)RB2、-N=S(O)RA2RB2、-S(O)2ORA2、-OS(O)2RA2、-NRA2S(O)rRB2、-NRA2S(O)(=NRE2)RB2、-S(O)rNRA2RB2、-S(O)(=NRE2)NRA2RB2、-NRA2S(O)2NRA2RB2、-NRA2S(O)(=NRE2)NRA2RB2、-P(O)RA2RB2and-P (O) (OR)A2)(ORB2) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
each RA1、RA2、RB1And RB2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RXSubstituted with the substituent(s);
or each "RA1And RB1"or" RA2And RB2Taken together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may be unsubstituted or substituted with 1,2 or 3 heteroatoms selected from RXSubstituted with the substituent(s);
each RE1And RE2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、ORa1、SRa1、-S(O)rRa1、-C(O)Ra1、C(O)ORa1、-C(O)NRa1Rb1and-S (O)rNRa1Rb1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or independently selected by at least one RYSubstituted with the substituent(s);
each RXIndependently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, -NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=N-ORb1)Ra1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tOC(O)Rb1、-(CRc1Rd1)tC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(O)rRb1、-(CRc1Rd1)tS(O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(O)Ra1Rb1、-(CRc1Rd1)tS(O)2ORb1、-(CRc1Rd1)tOS(O)2Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)Rb1、-(CRc1Rd1)tS(O)rNRa1Rb1、-(CRc1Rd1)tS(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)2NRa1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tP(O)Ra1Rb1And- (CR)c1Rd1)tP(O)(ORa1)(ORb1) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or independently selected by at least one RYSubstituted with the substituent(s);
each Ra1And Rb1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RYSubstituted with the substituent(s);
or Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Rc1And Rd1Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RYSubstituted with the substituent(s);
or Rc1And Rd1Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Re1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, CN, NO2、-ORa2、-SRa2、-S(O)rRa2、-C(O)Ra2、-C(O)ORa2、-S(O)rNRa2Rb2and-C (O) NRa2Rb2
Each RYIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tC(=NRe2)Ra2、-(CRc2Rd2)tC(=N-ORb2)Ra2、-(CRc2Rd2)tC(O)ORb2、-(CRc2Rd2)tOC(O)Rb2、-(CRc2Rd2)tC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)Rb2、-(CRc2Rd2)tC(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)Rb2、-(CRc2Rd2)tOC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)ORb2、-(CRc2Rd2)tNRa2C(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(S)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)NRa2Rb2、-(CRc2Rd2)tS(O)rRb2、-(CRc2Rd2)tS(O)(=NRe2)Rb2、-(CRc2Rd2)tN=S(O)Ra2Rb2、-(CRc2Rd2)tS(O)2ORb2、-(CRc2Rd2)tOS(O)2Rb2、-(CRc2Rd2)tNRa2S(O)rRb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)Rb2、-(CRc2Rd2)tS(O)rNRa2Rb2、-(CRc2Rd2)tS(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2S(O)2NRa2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tP(O)Ra2Rb2And- (CR)c2Rd2)tP(O)(ORa2)(ORb2) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from hydroxy, CN, amino, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Ra2And Rb2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, each of which is alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylaminoThe heterocyclyl, aryl and heteroaryl groups are unsubstituted or substituted by at least one member independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Ra2And Rb2Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted by 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Rc2And Rd2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Rc2And Rd2Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be substituted by 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Re2Independently selected from hydrogen, deuterium, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, -C (O) C1-4Alkyl, -C (O) C3-10Cycloalkyl, -C (O) OC1-4Alkyl, -C (O) OC3-10Cycloalkyl, -C (O) N (C)1-4Alkyl radical)2、-C(O)N(C3-10Cycloalkyl radicals2、-S(O)2C1-4Alkyl, -S (O)2C3-10Cycloalkyl, -S (O)2N(C1-4Alkyl radical)2and-S (O)2N(C3-10Cycloalkyl radicals2
n is selected from 0, 1,2,3 and 4;
each r is independently selected from 0, 1 and 2;
each t is independently selected from 0, 1,2,3 and 4;
the compounds provided do not include:
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-oxadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-oxadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-thiadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hexan-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) isoindolin-1-one,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5- (trifluoromethyl) -1,3, 4-thiadiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2-isopropylthiazole,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2- (trifluoromethyl) thiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropylthiazole, and
5-cyclopropyl-2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I), or at least one pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In another aspect, the present invention provides a method for modulating TRK, the method comprising administering to a system or subject in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, or a pharmaceutical composition thereof, thereby modulating TRK.
The present invention also provides a method of treating, ameliorating or preventing a condition responsive to inhibition of TRK comprising administering to a system or subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, and optionally in combination with a second therapeutic agent, to treat the condition.
Alternatively, the present invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a TRK-mediated condition. In particular embodiments, the compounds can be used alone or in combination with a second therapeutic agent to treat a TRK-mediated condition.
Alternatively, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment of a TRK-mediated condition.
In particular, wherein the disorder includes, but is not limited to, an autoimmune disease, a transplant disease, an infectious disease, or an abnormal cell proliferation.
In addition, the present invention provides a method of treating a cell proliferative disorder comprising administering to a system or subject in need thereof an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or pharmaceutical composition thereof, optionally in combination with a second therapeutic agent, to treat the disorder.
Alternatively, the present invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a disorder of abnormal cell proliferation. In particular embodiments, the compounds may be used alone or in combination with chemotherapeutic agents to treat the above-mentioned disorders.
In particular, wherein the disorder includes, but is not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of the breast, kidney, prostate, colorectal, thyroid, ovary, pancreas, neuron, lung, uterus or gastrointestinal tract.
In the above methods of using the compounds of the present invention, a compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered to a system comprising cells or tissues, or to an individual including a mammalian individual, such as a human or animal individual.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this patent belongs. All patents, patent applications, published publications, etc. referred to herein are incorporated by reference in their entirety unless otherwise indicated. As used in this patent, the same terms are defined differently than the definitions in this section.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any claims. In this application, the use of the singular includes the plural unless specifically stated otherwise. It is noted that, in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It is also noted that "or" represents "and/or" unless stated otherwise. Furthermore, "comprising," "including," and like terms are not intended to be limiting.
Unless otherwise indicated, the conventional techniques of mass spectrometry, nuclear magnetic resonance, high performance liquid chromatography, infrared and ultraviolet/visible spectroscopy, and pharmacology used in this patent are prior art. Unless specifically defined, the nomenclature, laboratory procedures, and techniques involved in analytical chemistry, synthetic organic chemistry, pharmaceutical and pharmaceutical chemistry of this patent are those well known. Standard techniques are available for chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and administration, and treatment of patients. The reaction and purification techniques may be carried out with reference to the manufacturer's instructions, or with reference to known, commonly used techniques, or with reference to the methods described in this patent. The techniques and procedures described above can be performed using methods that are conventional and well known in the literature cited in this specification. In the specification, groups and substituents may be selected by one skilled in the art to form stable structures and compounds.
When a substituent is referred to by a formula, the substituents in the formula are written from left to right as they are from right to left. E.g. CH2O and OCH2The same is true.
"substituted" means that the hydrogen atom is replaced with a substituent. It is noted that substituents on a particular atom are constrained by their valence states.
The term "C" as used hereini-j"or" i-j member "means that the moiety has i-j carbon atoms or i-j atoms. For example, "C1-6By alkyl is meant that the alkyl group has 1 to 6 carbon atoms. Likewise, C3-10Cycloalkyl means that the cycloalkyl group has 3 to 10 carbon atoms.
When any variable (e.g., R) occurs more than one time on the structure of a compound, it is independently defined in each instance. Thus, for example, if a group is substituted with 0-2R, that group may optionally be substituted with up to two R, and R has an independent choice in each case. In addition, combinations of substituents and/or variants thereof are permitted only if such combinations would result in stable compounds.
"one or more" or "at least one" means one, two, three, four, five, six, seven, eight, nine or more.
Unless otherwise indicated, the term "hetero" refers to a heteroatom or a heteroatom group (i.e., a group containing a heteroatom), i.e., an atom other than carbon and hydrogen atoms or a group containing such atoms. Preferably, the heteroatoms are independently selected from O, N, S, P, and the like. In embodiments involving two or more heteroatoms, the two or more heteroatoms may be the same, or the two or more heteroatoms may be partially or fully different.
"alkyl", alone or in combination with other terms, refers to branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Unless otherwise indicated, "alkyl" means C1-10An alkyl group. For example, "C1-6C in alkyl1-6"refers to a straight or branched chain arrangement of 1,2,3,4, 5 or 6 carbon atoms. For example, "C1-8Alkyl "includes, but is not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, heptyl, and octyl.
"cycloalkyl" whether used alone or in combination with other terms, refers to a monocyclic or bridged hydrocarbon ring system. Monocyclic cycloalkyl groups contain 3 to 10 carbon atoms, no heteroatoms, no double bonds. Examples of monocyclic systems include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. A monocyclic ring may comprise one or two alkylene bridges, each comprising 1,2 or 3 carbon atoms, which are bonded to two non-adjacent carbon atoms of the ring system. Representative examples of bridged cycloalkane systems include, but are not limited to, bicyclo [3.1.1] heptane, bicyclo [2.2.1] heptane, bicyclo [2.2.2] octane, bicyclo [3.2.2] nonane, bicyclo [3.3.1] nonane, bicyclo [4.2.1] nonane, tricyclo [3.3.1.03,7] nonane, and tricyclo [3.3.1.13,7] decane (adamantane). The monocyclic and bridged hydrocarbon rings can be attached to the parent molecular moiety through any substitutable atom in the ring system.
"alkenyl", alone or in combination with other terms, refers to a nonaromatic, straight chain, branched or cyclic hydrocarbon radical containing from 2 to 10 carbon atoms and having at least one carbon-carbon double bond. In some embodiments, 1 carbon-carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present. Thus, "C2-6Alkenyl "means alkenyl containing 2 to 6 carbon atoms. Alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, 2-methylbutenyl, and cyclohexenyl. The straight, branched or cyclic portion of the alkenyl group may contain a double bond, and substituted alkenyl groups, if indicated, may be substituted.
"alkynyl", whether used alone or in combination with other terms, refers to a straight, branched or cyclic hydrocarbon radical containing from 2 to 10 carbon atoms and at least one carbon-carbon triple bond. In some embodiments, up to 3 carbon-carbon triple bonds may be present. Thus, "C2-6Alkynyl "refers to alkynyl groups containing 2-6 carbon atoms. Alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, 3-methylbutynyl, and the like. The straight, branched or cyclic portion of the alkynyl group may contain a triple bond, and a substituted alkynyl group, if indicated, may be substituted.
"halogen" means fluorine, chlorine, bromine, iodine.
"alkoxy", used alone or in combination with other terms, refers to an alkyl group attached to an oxygen atom by a single bond. The alkoxy group is attached to the molecule through an oxygen atom. Alkoxy groups may be represented as-O-alkyl. "C1-10Alkoxy "refers to an alkoxy group containing 1 to 10 carbon atoms, and may be a straight chain or a branched structure. Alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, pentyloxy, hexyloxy, and the like.
"Cycloalkoxy", used alone or in combination with other terms, means a cycloalkyl group attached by a single bond to an oxygen atom. The cycloalkoxy group is attached to the molecule through an oxygen atom. Cycloalkoxy can be represented as-O-cycloalkyl. "C3-10Cycloalkoxy "means a cycloalkoxy group containing 3 to 10 carbon atoms. Cycloalkoxy includes, but is not limited to, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and the like.
"alkylthio", used alone or in combination with other terms, refers to an alkyl group attached by a single bond to a sulfur atom. Alkylthio groups are attached to the molecule through a sulfur atom. Alkylthio groups may be represented by-S-alkyl. "C1-10Alkylthio "refers to an alkylthio group containing 1 to 10 carbon atoms and can be a straight chain or branched structure. Alkylthio includes, but is not limited to, methylthio, ethylthio, propylthio, isopropylthio, butylthio, hexylthio, and the like.
"Cycloalkylsulfanyl", used alone or in combination with other terms, means a cycloalkyl group attached by a single bond to a sulfur atom. The cycloalkylthio group is bonded to the molecule through a sulfur atom. The cycloalkylthio group may be represented as-S-cycloalkyl. ' C3-10Cycloalkylthio "means a cycloalkylthio group containing 3 to 10 carbon atoms. Cycloalkylthio groups include, but are not limited to, cyclopropylthio, cyclobutylthio, and cyclohexylthio, and the like.
"alkylamino", used alone or in combination with other terms, refers to an alkyl group attached to a nitrogen atom by a single bond. The alkylamino group is attached to another molecule through a nitrogen atom. Alkylamino may be represented as-NH (alkyl). "C1-10Alkylamino "refers to alkylamino groups containing 1 to 10 carbon atoms and can be straight or branched. Alkylamino includes, but is not limited to, methylamino, ethylamino, propylamino, isopropylamino, butylamino, hexylamino and the like.
"Cycloalkylamino", used alone or in combination with other terms, refers to a cycloalkyl group attached to a nitrogen atom by a single bond.The cycloalkylamino group is linked to another molecule through a nitrogen atom. The cycloalkylamino group may be represented as-NH (cycloalkyl). ' C3-10Cycloalkylamino "refers to cycloalkylamino groups containing 3 to 10 carbon atoms. Cycloalkylamino groups include, but are not limited to, cyclopropylamino, cyclobutylamino, cyclohexylamino and the like.
"Di (alkyl) amino", used alone or in combination with other terms, refers to two alkyl groups attached to a nitrogen atom by a single bond. The di (alkyl) amino group is attached to the molecule through a nitrogen atom. The di (alkyl) amino group may be represented by-N (alkyl)2. ' two (C)1-10Alkyl) amino "means a di (C) group in which the two alkyl moieties each contain 1 to 10 carbon atoms1-10Alkyl) amino, which may be linear or branched.
"aryl", used alone or in combination with other terms, includes: 5-and 6-membered aromatic carbocyclic rings, such as phenyl; bicyclic rings having at least one aromatic carbon ring, such as naphthyl, indane and 1,2,3, 4-tetrahydroquinoline, and tricyclic rings having at least one aromatic carbon ring, such as fluorene. Aryl substituents are considered to be linked through an aromatic ring if they are bicyclic or tricyclic and at least one of the rings is non-aromatic. An "aryl" group may contain 5 to 20 carbon atoms (C)5-20Aryl), for example: 6 to 14 carbon atoms (C)6-14Aryl) or 6 to 10 carbon atoms (C)6-10Aryl) such as phenyl, naphthyl, indanyl, fluorenyl, and the like.
For example, aryl includes 5-and 6-membered carbocyclic aromatic rings fused to a 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O, and S, provided that the attachment site is a carbocyclic aromatic ring. Divalent radicals, which are formed from substituted benzene derivatives and have free valence electrons present at the ring atoms, are designated as substituted phenylene radicals. Divalent radicals derived from monovalent polycyclic hydrocarbon radicals whose name ends with "-yl", which are obtained by removing one more hydrogen atom from a carbon atom containing a free valence electron, are named after the name of the monovalent radical plus "-idene (-idene)", for example, naphthyl, which has two attachment sites, is called naphthylidene. The definition of aryl, however, does not include, nor overlap with, heteroaryl, and is defined individually as follows. Thus, if one or more aromatic carbocyclic rings are fused to an aromatic ring of a heterocyclic ring, the resulting ring system should be considered heteroaryl as defined herein, rather than aryl.
"heteroaryl", used alone or in combination with other terms, means
A 5-to 8-membered aromatic monocyclic ring containing 1 to 4, in some cases 1, N, O and S
In the examples 1 to 3 heteroatoms, the remainder being carbon atoms;
an 8-to-12-membered bicyclic ring containing 1 to 4, and in certain embodiments 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms, and wherein at least one heteroatom is present in the aromatic ring
Performing the following steps; and
11-to 14-membered tricyclic rings. The ring contains 1 to 4, and in certain embodiments 1 to 3 heteroatoms selected from N, O and S, the remainder being carbon atoms, and at least one of the heteroatoms is present in the aromatic ring.
When the total number of S and O in the heteroaryl group is greater than 1, these heteroatoms are not adjacent to each other. In some embodiments, the total number of S and O in the heteroaryl group is no greater than 2. In some embodiments, the total number of S and O in the heteroaryl group is no greater than 1.
Examples of heteroaryl groups include, but are not limited to (the numbering of the attachment site is preferred, designating position 1) 2-pyridyl, 3-pyridyl, 4-pyridyl, 2, 3-pyrazinyl, 3, 4-pyrazinyl, 2, 4-pyrimidinyl, 3, 5-pyrimidinyl, 1-pyrazolyl, 2,3 pyrazolyl, 2, 4-imidazolinyl, isoxazolyl, oxazolyl, thiazolyl, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuranyl, benzimidazolyl, indolinyl, pyridazinyl, triazolyl, quinolyl, pyrazolyl and 5,6,7, 8-tetrahydroisoquinolyl.
Further, heteroaryl groups include, but are not limited to, pyrrolyl, isothiazolyl, triazinyl, pyrazinyl, pyridazinyl, indolyl, benzotriazolyl, quinoxalinyl and isoquinolyl. "heteroaryl" includes N-oxide derivatives of nitrogen-containing heteroaryl groups, as defined below for heterocyclic groups.
The nomenclature of a monovalent heteroaryl group ends with the "-yl", the name of a derived divalent group being obtained by removing a further hydrogen atom from the carbon atom containing the free valence electron, this divalent group being the name of a monovalent group plus the "-idene" — "group", for example: the pyridyl group having two attachment sites is called a pyridylidene. The definition of heteroaryl does not include, nor overlap with, aryl as defined above.
"heterocycle" (and derivatives thereof such as "heterocyclic" or "heterocyclyl") refers broadly to a single cyclic aliphatic hydrocarbon, typically having from 3 to 12 ring atoms, containing at least 2 carbon atoms, and further containing from 1 to 3 heteroatoms independently selected from oxygen, sulfur, nitrogen, and phosphorus, as well as combinations comprising at least one of the foregoing heteroatoms. Alternatively, the heterocyclic ring in the above definition may be a polycyclic ring system (e.g., bicyclic ring), wherein two or more rings may be linked by a fused, bridged or spiro ring, wherein at least one ring contains one or more heteroatoms independently selected from oxygen, sulfur, nitrogen, and phosphorus. "heterocyclic ring" also refers to 5-to 7-membered heterocyclic rings fused to 5-and 6-membered aromatic carbocyclic rings containing one or more heteroatoms selected from oxygen, sulfur, nitrogen, and phosphorus, provided that the site of attachment is on the heterocyclic ring. Heterocycles may be saturated or contain one to more double bonds (i.e., partially unsaturated). The heterocyclic ring may be substituted with oxo (oxo). Either the carbon atom or the heteroatom of the heterocycle may be the attachment site, provided that a stable structure is formed. When a substituent is present on the heterocycle, the substituent may be attached to any heteroatom or carbon atom on the heterocycle, provided that a stable chemical structure is formed. The heterocyclic and heteroaryl definitions described herein do not overlap.
Suitable heterocycles include, for example (attachment site is preferably ordered 1) 1-pyrrolidinyl, 2, 4-imidazolidinyl, 2, 3-pyrazolidinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2, 5-piperazinyl, 1, 4-piperazinyl and 2, 3-pyridazinyl. Also contemplated are morpholinyl groups, including 2-morpholinyl and 3-morpholinyl (oxygen atom position numbering is preferably 1). Substituted heterocycles also include ring systems substituted with one or more oxo groups, such as piperidinyl-N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl, and 1, 1-dioxo-1-thiomorpholinyl. Bicyclic heterocycles include, but are not limited to:
Figure BDA0003541883110000111
as used herein, "aryl-alkyl" refers to an aryl-substituted alkyl group. Exemplary aralkyl groups include benzyl, phenethyl, naphthylmethyl, and the like. In some embodiments, aralkyl contains 7 to 20 or 7 to 11 carbon atoms. When using "aryl radicals C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl portion rather than the aryl portion.
As used herein, "heterocyclyl-alkyl" refers to a heterocyclyl-substituted alkyl group. When using "heterocyclyl C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl moiety rather than in the heterocyclyl moiety.
As used herein, "cycloalkyl-alkyl" refers to cycloalkyl-substituted alkyl. When using "C3-10cycloalkyl-C1-4Alkyl "in which" C3-10"refers to the number of carbon atoms in the cycloalkyl moiety rather than the alkyl moiety. Wherein "C1-4"refers to the number of carbon atoms in the alkyl moiety rather than the cycloalkyl moiety.
As used herein, "heteroaryl-alkyl" refers to heteroaryl-substituted alkyl. When using "heteroaryl-C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl moiety rather than in the heteroaryl moiety.
To avoid ambiguity, for example: when alkyl, cycloalkyl, heterocyclylalkyl, aryl, and/or heteroaryl substituents thereof are mentioned, it is meant that each of these groups is substituted individually or that these groups are mixed. That is: if R is1Is aryl-C1-4The alkyl, aryl moieties may be unsubstituted or substituted by at least one, such as 1,2,3 or 4 independently selected from RXMay also be unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXA substituent of (1).
"pharmaceutically acceptable salt" refers to a salt with a pharmaceutically acceptable non-toxic base or acid, including inorganic or organic bases and inorganic or organic acids. The salt of an inorganic base may be selected, for example, from: aluminum, ammonium, calcium, copper, iron, ferrous iron, lithium, magnesium, manganese, manganous, potassium, sodium and zinc salts. Further, the pharmaceutically acceptable salt of inorganic base may be selected from ammonium, calcium, magnesium, potassium, sodium salt. One or more crystal structures may be present in the solid salt, as well as in the form of hydrates. The pharmaceutically acceptable salts of organic non-toxic bases may be selected, for example, from: primary, secondary and tertiary amine salts, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, such as arginine, betaine, caffeine, choline, N' -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine.
When the compound referred to in this patent is a base, it is necessary to prepare a salt thereof with at least one pharmaceutically acceptable non-toxic acid selected from inorganic acids and organic acids. For example, selected from acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, pamoic acid, pantothenic acid, phosphoric acid, succinic acid, sulfuric acid, tartaric acid, p-toluenesulfonic acid. In some embodiments, these acids may be selected, for example: citric acid, hydrobromic acid, hydrochloric acid, maleic acid, phosphoric acid, sulfuric acid, fumaric acid, tartaric acid.
By "administering" or "administration" of a compound or a pharmaceutically acceptable salt thereof is meant providing a compound of the invention or a pharmaceutically acceptable salt thereof to a subject in need of treatment.
An "effective amount" is an amount of a compound or a pharmaceutically acceptable salt thereof that is capable of eliciting a biological or medical response in a tissue, system, animal or human that is observable by a researcher, veterinarian, clinician or other clinician.
"composition" comprising: the invention may take the form of a kit, article of manufacture, or any combination thereof. A pharmaceutical composition comprising: products comprising the active ingredient and an inert ingredient as a carrier, as well as products produced by any two or more of the ingredients, directly or indirectly, by combination, complexation or aggregation, or by dissociation of one or more of the ingredients, or by other types of reactions or interactions of one or more of the ingredients.
By "pharmaceutically acceptable" is meant compatible with the other ingredients of the formulation and not unacceptably toxic to the user.
"subject" refers to subjects suffering from a disease, disorder, or the like, and includes mammals and non-mammals. Mammals include, but are not limited to, any member of the mammalian family: humans, non-human primates such as chimpanzees, and other apes and monkeys; farm animals such as cattle, horses, sheep's yang, goats, pigs; domestic animals such as rabbits, dogs, and cats; the experimental animals include rodents such as rats, mice, guinea pigs, and the like. Non-mammalian animals include, but are not limited to, birds, fish, and the like. In one embodiment of the invention, the mammal is a human.
"treating" includes alleviating, alleviating or ameliorating a disease or condition, preventing other conditions, ameliorating or preventing a metabolic factor underlying a condition, inhibiting a disease or condition, e.g., arresting the development of a disease or condition, alleviating a disease or condition, promoting remission of a disease or condition, or arresting the signs of a disease or condition, and extends to include prevention. "treating" also includes achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit refers to eradication or amelioration of the condition being treated. In addition, therapeutic benefit is achieved by eradicating or ameliorating one or more physiological signs associated with the underlying disease, and amelioration of the disease in the patient is observed, although the patient may still be suffering from the underlying disease. Prophylactic benefit refers to the use of a composition by a patient to prevent the risk of a disease, or the use of a patient presenting with one or more physiological conditions of a disease, although the disease has not yet been diagnosed.
"protecting group" (Pg) refers to a class of substituents used to block or protect a particular functional group by reacting with other functional groups on a compound. For example, "amino protecting group" refers to a substituent attached to an amino group that blocks or protects the amino functionality on a compound. Suitable amino protecting groups include acetyl, trifluoroacetyl, t-Butyloxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and the 9-fluorenylmethoxycarbonyl protecting group (Fmoc). Similarly, "hydroxy protecting group" refers to a class of hydroxy substituents that are effective in blocking or protecting the hydroxy function. Suitable protecting groups include, but are not limited to, acetyl and silyl groups. "carboxy protecting group" refers to a class of carboxy substituents that function effectively to block or protect a carboxy group. Common carboxyl protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfinyl) ethyl, 2- (diphenylphosphino) -ethyl, nitroethyl and the like. For general description and instructions for use of protecting groups, see references: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991。
"NH protecting group" includes, but is not limited to, trichloroethoxycarbonyl, tribromoethoxycarbonyl, benzyloxycarbonyl, p-nitrobenzoyl, o-bromobenzyloxycarbonyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetyl, phenylacetyl, formyl, acetyl, benzoyl, t-pentyloxycarbonyl, t-butyloxycarbonyl, p-methoxybenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, 4- (phenylazo) benzyloxycarbonyl, 2-furfuryloxycarbonyl, diphenylmethoxycarbonyl, 1-dimethylpropoxycarbonyl, isopropyloxycarbonyl, phthaloyl, succinyl, alanyl, leucyl, 1-adamantyloxycarbonyl, 8-quinolinyloxycarbonyl, benzyl, benzhydryl, trityl, 2-nitrobenzenesulfonyl, methylsulfonyl, p-toluenesulfonyl, and the like, N, N-dimethylaminomethylene, benzylidene, 2-hydroxybenzylidene, 2-hydroxy-5-chlorobenzylidene, 2-hydroxy-l-naphthylmethylene, 3-hydroxy-4-pyridylmethylidene, cyclohexylidene, 2-ethoxycarbonylcyclohexylidene, 2-ethoxycarbonylcyclopentylidene, 2-acetylcyclohexylidene, 3-dimethyl-5-oxocyclohexylidene, diphenylphosphoryl, dibenzylphosphoryl, 5-methyl-2-oxo-2H-l, 3-dioxol-4-yl-methyl, trimethylsilyl, triethylsilyl and triphenylsilyl.
"C (O) OH" protecting groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, 1-dimethylpropyl, n-butyl, t-butyl, phenyl, naphthyl, benzyl, benzhydryl, trityl, p-nitrobenzyl, p-methoxybenzyl, bis (p-methoxyphenyl) methyl, acetylmethyl, phenacyl, p-nitrobenzoylmethyl, p-bromobenzoylmethyl, p-methanesulfonylbenzoylmethyl, 2-tetrahydropyranyl, 2-tetrahydrofuranyl, 2,2, 2-trichloroethyl, 2- (trimethylsilyl) ethyl, acetoxymethyl, propionyloxymethyl, pivaloyloxymethyl, phthalimidomethyl, succinimidylmethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxymethyl, methoxyethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, Benzyloxymethyl, methylthiomethyl, 2-methylthioethyl, phenylthiomethyl, 1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, trimethylsilyl, triethylsilyl, triisopropylsilyl, diethylisopropylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl and tert-butylmethoxyphenylsilyl.
"OH or SH" protecting groups include, but are not limited to, benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4-bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, methoxycarbonyl, ethoxycarbonyl, t-butoxycarbonyl, 1-dimethylpropoxycarbonyl, isopropoxycarbonyl, isobutoxycarbonyl, diphenylmethoxycarbonyl, 2,2, 2-trichloroethoxycarbonyl, 2,2, 2-tribromoethoxycarbonyl, 2- (trimethylsilane) ethoxycarbonyl, 2- (phenylsulfonyl) ethoxycarbonyl, 2- (triphenylphosphonio) ethoxycarbonyl, 2-furfuryloxycarbonyl, 1-adamantyloxycarbonyl, vinyloxycarbonyl, allyloxycarbonyl, 4-ethoxy-1-naphthyloxycarbonyl, N-vinyloxycarbonyl, N-type-carbonyl, N-O-1-naphthyloxycarbonyl, N-carbonyl, N-vinyloxycarbonyl, N-O-carbonyl, N-O-carbonyl, 8-quinolyloxycarbonyl, acetyl, formyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, pivaloyl, benzoyl, methyl, tert-butyl, 2,2, 2-trichloroethyl, 2-trimethylsilylethyl, 1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, benzyl (phenylmethyl), p-methoxybenzyl, 3, 4-dimethoxybenzyl, diphenylmethyl, triphenylmethyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-methoxyethoxymethyl, 2,2, 2-trichloroethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, dichloroacetyl, benzoyl, chloroacetyl, trichloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, pivaloyl, benzoyl, benzyl (phenylmethyl), p-methoxybenzyl, 3, 4-dimethoxybenzyl, diphenylmethyl, triphenylmethyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2- (trimethylsilyl) ethoxymethyl, 2-ethyl, ethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, 2-ethyl, 2-ethoxymethyl, 2- (trimethylsilyl) methyl, 2-ethoxymethyl, 2-methyl, 2-n, and a, 1-ethoxyethyl group, methanesulfonyl group, p-toluenesulfonyl group, trimethylsilyl group, triethylsilyl group, triisopropylsilyl group, diethylisopropylsilyl group, tert-butyldimethylsilyl group, tert-butyldiphenylsilyl group, diphenylmethylsilyl group and tert-butylmethoxyphenylsilyl group.
Geometric isomers may exist in the compounds of the present invention. Compounds of the present invention may have carbon-carbon double bonds or carbon-nitrogen double bonds in either the E or Z configuration, where "E" represents the preferred substituent on the opposite side of the carbon-carbon double bond or carbon-nitrogen double bond and "Z" represents the preferred substituent on the same side of the carbon-carbon double bond or carbon-nitrogen double bond, as defined by Cahn-Ingold-Prelog preference. The compounds of the invention may also exist as mixtures of "E" and "Z" isomers. The substituents around the cycloalkyl or heterocyclyl group may be in either the cis or trans configuration. In addition, the present invention includes different isomers and mixtures thereof formed by different arrangements of substituents around the adamantane ring system. Two substituents around a single ring in an adamantane ring system are designated in either the Z or E relative configuration. See, for example, C.D.Jones, M.Kaselj, R.N.Salvatore, W.J.le Noble J.org.chem.1998,63, 2758-.
Compounds of the invention may contain asymmetrically substituted carbon atoms of R or S configuration, "R" and "S" are defined in IUPAC 1974Recommendations for Section E, functional Stereochemistry, Pure appl. chem. (1976)45, 13-10. Compounds containing asymmetrically substituted carbon atoms are racemates if the amounts of R and S configuration are the same. If one of the configurations is present in greater amounts than the other, the configuration of the chiral carbon atom is represented by the more abundant configuration, preferably with an enantiomeric excess of about 85-90%, more preferably about 95-99%, and even more preferably about 99% or more. Thus, the present invention encompasses racemic mixtures, relative and absolute stereoisomers, and mixtures of relative and absolute stereoisomers.
Isotopically enriched or labelled compounds
The compounds of the invention may exist in isotopically-labelled or enriched forms containing one or more atoms of different mass and mass numbers from the atom mass and mass number most prevalent in nature. The isotope may be a radioactive or non-radioactive isotope. Isotopes of atoms such as hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine include, but are not limited to,2H、3H、13C、14C、15N、18O、32P、35S、18F、36cl and125I. other isotopes and/or other atoms containing these atoms are also within the scope of the invention.
In another embodiment, the isotopically labeled compound comprises deuterium (A), (B), (C) and D) and (C)2H) Tritium (a)3H) Or14Isotope of C. Isotopically-labelled compounds of the present invention can be obtained by methods well known to those skilled in the art. These isotopically labeled compounds can be obtained by substituting a non-labeling reagent with an isotopically labeled reagent by referring to the examples and reaction schemes of the present invention. In certain examples, the compound can be treated with an isotopic tagging agent to replace an atom with an isotopic atom, e.g., replacement of a hydrogen with a deuterium can be accomplished by a deuterated acid such as D2SO4/D2And exchanging the action of O.
The isotope labeled compound of the invention can be used as the standard of the TRK inhibitor drug effect combination test. Isotopically-containing compounds are useful in pharmaceutical studies to evaluate the mechanism of action and metabolic pathways of non-isotopically labeled parent compounds, to study the in vivo metabolic turnover of compounds (Blake et al.J.Pharm.Sci.64,3,367-391 (1975)). Such metabolic studies are important for the design of safe and effective therapeutic agents, and can be judged to be toxic or carcinogenic to the in vivo active compound administered to the patient or to the metabolite of the parent compound (Foster et al, Advances in Drug Research Vol.14, pp.2-36, Academic press, London, 1985; Kato et al, J.Labelled Comp.Radiopharmaceut.,36(10):927-932 (1995); Kushner et al, Can.J.Physiol.Pharmacol,77,79-88 (1999)).
In addition, drugs containing non-reflex active isotopes, such as deuterated drugs, known as "heavy drugs," are useful for treating diseases and disorders associated with TRK activity. The proportion of a certain isotope in a compound that exceeds its natural abundance is called enrichment. The amount of enrichment is, for example, from about 0.5, 1,2,3,4, 5,6,7,8, 9, 10, 12, 16, 21, 25, 29, 33, 37, 42, 46, 50, 54, 58, 63, 67, 71, 75, 79, 84, 88, 92, 96 to about 100 mol%.
Drug-stable isotopic labeling can alter its physicochemical properties, such as pKa and liquid solubility. If isotopic substitution affects the region associated with ligand-receptor interaction, then these effects and changes may affect the pharmacodynamic response of the drug molecule. Certain physical properties of stable isotope-labeled molecules differ from those of unlabeled molecules, while chemical and biological properties are the same, but with one important difference: any chemical bond containing a heavy isotope and another atom is stronger than a light isotope due to the increased mass of the heavy isotope. Accordingly, the presence of isotopes at the metabolic or enzymatic conversion sites slows the reaction and may alter its pharmacokinetic or pharmacodynamic properties compared to non-isotopically labeled compounds.
In embodiment (1), the present invention provides a compound of formula (I),
Figure BDA0003541883110000161
or a pharmaceutically acceptable salt thereof, wherein:
when X is N, Y is C, as shown in formula (Ia);
Figure BDA0003541883110000162
when X is C, Y is N, as shown in formula (Ib);
Figure BDA0003541883110000163
R1selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with a substituent of (1);
each R2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA1RB1、-ORA1-C(O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(O)ORA1、-OC(O)RA1、-C(O)NRA1RB1、-NRA1C(O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(O)NRA1RB1、-NRA1C(O)ORB1、-NRA1C(O)NRA1RB1、-NRA1C(S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(O)rRA1、-S(O)(=NRE1)RB1、-N=S(O)RA1RB1、-S(O)2ORA1、-OS(O)2RA1、-NRA1S(O)rRB1、-NRA1S(O)(=NRE1)RB1、-S(O)rNRA1RB1、-S(O)(=NRE1)NRA1RB1、-NRA1S(O)2NRA1RB1、-NRA1S(O)(=NRE1)NRA1RB1、-P(O)RA1RB1and-P (O) (OR)A1)(ORB1) Wherein each alkyl, alkenyl, alkynyl,The cycloalkyl, heterocyclyl, aryl and heteroaryl are unsubstituted or substituted with at least one substituent independently selected from RXSubstituted with the substituent(s);
R3selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
R4selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA2RB2、-ORA2、-C(O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(O)ORA2、-OC(O)RA2、-C(O)NRA2RB2、-NRA2C(O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(O)NRA2RB2、-NRA2C(O)ORB2、-NRA2C(O)NRA2RB2、-NRA2C(S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(O)rRA2、-S(O)(=NRE2)RB2、-N=S(O)RA2RB2、-S(O)2ORA2、-OS(O)2RA2、-NRA2S(O)rRB2、-NRA2S(O)(=NRE2)RB2、-S(O)rNRA2RB2、-S(O)(=NRE2)NRA2RB2、-NRA2S(O)2NRA2RB2、-NRA2S(O)(=NRE2)NRA2RB2、-P(O)RA2RB2and-P (O) (OR)A2)(ORB2) Wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least oneOne is independently selected from RXSubstituted with the substituent(s);
each RA1、RA2、RB1And RB2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RXSubstituted with a substituent of (1);
or each "RA1And RB1"or" RA2And RB2Taken together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may be unsubstituted or substituted with 1,2 or 3 heteroatoms selected from RXSubstituted with the substituent(s);
each RE1And RE2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、ORa1、SRa1、-S(O)rRa1、-C(O)Ra1、C(O)ORa1、-C(O)NRa1Rb1and-S (O)rNRa1Rb1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RYSubstituted with the substituent(s);
each RXIndependently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, arylRadical, aryl radical-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, -NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=N-ORb1)Ra1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tOC(O)Rb1、-(CRc1Rd1)tC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(O)rRb1、-(CRc1Rd1)tS(O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(O)Ra1Rb1、-(CRc1Rd1)tS(O)2ORb1、-(CRc1Rd1)tOS(O)2Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)Rb1、-(CRc1Rd1)tS(O)rNRa1Rb1、-(CRc1Rd1)tS(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)2NRa1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tP(O)Ra1Rb1And- (CR)c1Rd1)tP(O)(ORa1)(ORb1) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RYSubstituted with a substituent of (1);
each Ra1And Rb1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RYSubstituted with the substituent(s);
or Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Rc1And Rd1Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least oneOne is independently selected from RYSubstituted with the substituent(s);
or Rc1And Rd1Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Re1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl, CN, NO2、-ORa2、-SRa2、-S(O)rRa2、-C(O)Ra2、-C(O)ORa2、-S(O)rNRa2Rb2and-C (O) NRa2Rb2
Each RYIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tC(=NRe2)Ra2、-(CRc2Rd2)tC(=N-ORb2)Ra2、-(CRc2Rd2)tC(O)ORb2、-(CRc2Rd2)tOC(O)Rb2、-(CRc2Rd2)tC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)Rb2、-(CRc2Rd2)tC(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)Rb2、-(CRc2Rd2)tOC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)ORb2、-(CRc2Rd2)tNRa2C(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(S)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)NRa2Rb2、-(CRc2Rd2)tS(O)rRb2、-(CRc2Rd2)tS(O)(=NRe2)Rb2、-(CRc2Rd2)tN=S(O)Ra2Rb2、-(CRc2Rd2)tS(O)2ORb2、-(CRc2Rd2)tOS(O)2Rb2、-(CRc2Rd2)tNRa2S(O)rRb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)Rb2、-(CRc2Rd2)tS(O)rNRa2Rb2、-(CRc2Rd2)tS(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2S(O)2NRa2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tP(O)Ra2Rb2And- (CR)c2Rd2)tP(O)(ORa2)(ORb2) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from hydroxy, CN, amino, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Ra2And Rb2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Ra2And Rb2Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Rc2And Rd2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Rc2And Rd2Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be substituted by 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Re2Independently selected from hydrogen, deuterium, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, -C (O) C1-4Alkyl, -C (O) C3-10Cycloalkyl, -C (O) OC1-4Alkyl, -C (O) OC3-10Cycloalkyl, -C (O) N (C)1-4Alkyl radical)2、-C(O)N(C3-10Cycloalkyl radicals2、-S(O)2C1-4Alkyl, -S (O)2C3-10Cycloalkyl, -S (O)2N(C1-4Alkyl radical)2and-S (O)2N(C3-10Cycloalkyl radicals2
n is selected from 0, 1,2,3 and 4;
each r is independently selected from 0, 1 and 2;
each t is independently selected from 0, 1,2,3, and 4;
the compounds provided do not include:
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-oxadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hexan-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-oxadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-thiadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hexan-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) isoindolin-1-one,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5- (trifluoromethyl) -1,3, 4-thiadiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2-isopropylthiazole,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2- (trifluoromethyl) thiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropylthiazole, and
5-cyclopropyl-2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole.
In another embodiment (2), the present invention provides a compound of embodiment (1), or a pharmaceutically acceptable salt thereof, wherein X is N, Y is C, the compound is represented by (Ia),
Figure BDA0003541883110000201
wherein R is1、R2、R3、R4And n is as defined for formula (I).
In another embodiment (3), the present invention provides a compound of embodiment (1), or a pharmaceutically acceptable salt thereof, wherein X is C, Y is N, the compound is represented by (Ib),
Figure BDA0003541883110000202
wherein R is1、R2、R3、R4And n is as defined for formula (I).
In another embodiment (4), the present invention provides a compound of any one of embodiments (1) - (3), or a pharmaceutically acceptable salt thereof, wherein R3Selected from 5-membered rings.
In another embodiment (5), the present invention provides a compound of any one of embodiments (1) - (3), or a pharmaceutically acceptable salt thereof, wherein R is3Selected from 6 membered rings.
In another embodiment (6), the present invention provides a compound of embodiment (4) or a pharmaceutically acceptable salt thereof, wherein R is3Is selected from
Figure BDA0003541883110000211
Figure BDA0003541883110000212
Which is unsubstituted or substituted by at least one member independently selected from RXIs substituted with the substituent(s).
In another embodiment (7), the present invention provides a compound of embodiment (5) or a pharmaceutically acceptable salt thereof, wherein R3Is selected from
Figure BDA0003541883110000213
Which is unsubstituted or substituted by at least one member independently selected from RXIs substituted with the substituent(s).
In another embodiment (8), the present invention provides a compound of any one of embodiments (1) - (7), or a pharmaceutically acceptable salt thereof, wherein R3Substituent R ofXSelected from halogen, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl, heterocyclyl-C1-4Alkyl, - (CR)c1Rd1)tORb1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(O)Ra1And- (CR)c1Rd1)tN=S(O)Ra1Rb1Wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from RYIs substituted with the substituent(s).
In another embodiment (9), the present invention provides a compound of embodiment (8), or a pharmaceutically acceptable salt thereof, wherein each R isa1And each Rb1Independent selectionFrom hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl and heterocyclyl, wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from RYIs substituted with the substituent(s).
In another embodiment (10), the present invention provides a compound of embodiment (8) or a pharmaceutically acceptable salt thereof, wherein Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-to 10-membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1,2 or 3RYAnd (4) substituting the group.
In another embodiment (11), the present invention provides a compound of embodiment (8), or a pharmaceutically acceptable salt thereof, wherein t is independently selected from 0, 1,2,3, each R is independently selected fromc1And each Rc1Independently selected from hydrogen, halogen, C1-10Alkyl radical, C3-10Cycloalkyl and heterocyclyl, wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one substituent independently selected from RYIs substituted with the substituent(s).
In another embodiment (12), the present invention provides a compound of embodiment (8), or a pharmaceutically acceptable salt thereof, wherein RYIs selected from C1-10Alkyl radical, C3-10Cycloalkyl, heterocyclyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tS(O)rRb2And- (CR)c2Rd2)tORb2Wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from OH, CN, amino, halogen, C1-10Alkyl and C1-10Alkoxy substituent.
In another embodiment (13), the present invention provides a compound of embodiment (12), or a pharmaceutically acceptable salt thereof, wherein each R isa2And each Rb2Independently selected from hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl and heterocyclyl, wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted by at least one group independently selected from OH, CN, amino, halogen, C1-10Alkyl and C1-10Substituent of alkoxy.
In another embodiment (14), the present invention provides a compound of embodiment (12), or a pharmaceutically acceptable salt thereof, wherein t is independently selected from 0, 1,2,3, each R is independently selected fromc2And each Rd2Independently selected from hydrogen, halogen, C1-10Alkyl radical, C3-10Cycloalkyl and heterocyclyl, wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from OH, CN, amino, halogen, C1-10Alkyl and C1-10Substituent of alkoxy.
In another embodiment (15), the present invention provides a compound of any one of embodiments (1) - (14), or a pharmaceutically acceptable salt thereof, wherein R3Substituent R ofXIs selected from-NH2、-CN、-CO2ET, methyl, isopropyl, cyclopropyl,
Figure BDA0003541883110000221
In another embodiment (16), the present invention provides a compound of any one of embodiments (1) - (15), or a pharmaceutically acceptable salt thereof, wherein R1Is aryl, wherein aryl is unsubstituted or substituted with at least one group independently selected from RXIs substituted.
In another embodiment (17), the present invention provides a compound of embodiment (16), or a pharmaceutically acceptable salt thereof, wherein R1Is phenyl, wherein phenyl is substituted by halogen, preferably, R1Is that
Figure BDA0003541883110000222
In another embodiment (18), the present invention provides a compound of any one of embodiments (1) - (17), or a pharmaceutically acceptable salt thereof, wherein each R is2Is independently selected from C1-10Alkyl radical, C3-10Cycloalkyl and halogen, wherein each alkyl and cycloalkyl is unsubstituted or substituted with at least one group independently selected from RXIs substituted with the substituent(s).
In another embodiment (19), the present invention provides a compound of any one of embodiments (1) - (17), or a pharmaceutically acceptable salt thereof, wherein n is selected from 0 and 1.
In another embodiment (20), the present invention provides a compound of any one of embodiments (1) - (19), or a pharmaceutically acceptable salt thereof, wherein R4Selected from hydrogen, halogen, -CN, -NRA2RB2and-C (O) NRA2RB2
In another embodiment (21), the present invention provides a compound of embodiment (20), or a pharmaceutically acceptable salt thereof, wherein R4Selected from hydrogen, Br, -NH2-CN and-C (O) NH2
In another embodiment (22), the compounds provided herein are selected from:
Figure BDA0003541883110000231
Figure BDA0003541883110000241
Figure BDA0003541883110000251
Figure BDA0003541883110000261
and pharmaceutically acceptable salts thereof.
In another embodiment (23), the compounds provided herein are selected from:
Figure BDA0003541883110000262
Figure BDA0003541883110000271
Figure BDA0003541883110000281
Figure BDA0003541883110000291
and pharmaceutically acceptable salts thereof.
In another embodiment (24), the present invention provides a pharmaceutical composition comprising a compound of any one of embodiments (1) - (23), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
In another embodiment (25), the invention provides a method of treating, ameliorating or preventing a condition responsive to inhibition of TRK comprising administering to a subject in need thereof an effective amount of a compound of any one of embodiments (1) - (23), or a pharmaceutically acceptable salt thereof, or at least one pharmaceutical composition thereof, optionally in combination with a second therapeutic agent.
In another embodiment (26), the invention provides the use of a compound of any one of embodiments (1) - (23), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a TRK-mediated disorder.
In another embodiment (27), the invention provides the use of embodiment (26), wherein the condition includes, but is not limited to, an autoimmune disease, a transplant disease, pain, an infectious disease, or a cell proliferative disease.
In another embodiment (28), the invention provides the use of embodiment (27), wherein the conditions include, but are not limited to, brain, lung, squamous cell, bladder, stomach, pancreas, breast, head, neck, renal region, kidney, ovary, prostate, colorectal, epidermal, esophageal, testicular, gynecological or thyroid cancer, benign hyperplasia of the skin, restenosis and benign prostatic hypertrophy, pancreatitis, kidney disease, chronic and/or acute pain; preventing blastocyst implantation, psoriasis, rashes and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, atherosclerosis, inhibition of keratinocytes responsive to a mixture of growth factors, Chronic Obstructive Pulmonary Disease (COPD) and other diseases.
In another aspect, the invention provides a kit comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof; and instructions including one or more of the following: information on what disease state the ingredient applies to, information stored on the ingredient, dosage information, and instructions on how to use the ingredient. In one particular variant, the kit comprises the compound in a multiple dose form.
In another aspect, the present invention provides an article of manufacture comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof; and a packaging material. In one variation, the packaging material comprises a container. In one particular variation, the container includes a label that identifies one or more of the following: instructions for what disease state the compound is to be administered, stored information, dosage information, and/or how to administer the compound. In another variation, the article of manufacture comprises the compound in a multiple dose form.
In another aspect, the invention provides a method of treatment comprising administering to a subject a compound disclosed herein, or a pharmaceutically acceptable salt thereof.
In another aspect, the present invention provides a method of inhibiting TRK by contacting a compound disclosed herein, or a pharmaceutically acceptable salt thereof, with TRK.
In another aspect, the present invention provides a method of inhibiting TRK comprising causing a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to be present in a subject to inhibit TRK activity in vivo.
In another aspect, the invention provides a method of inhibiting TRK comprising administering to a subject a first compound that converts to a second compound in vivo, wherein the second compound inhibits TRK activity in vivo, and the second compound is a compound or variant of any of the above embodiments.
In another aspect, the invention provides a method of treating a disease state in which TRK activity contributes to the pathology and/or symptomology of the disease state, the method comprising causing a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to be present in a subject.
In another aspect, the invention provides a method of treating a disease state for which TRK activity contributes to the pathology and/or symptomology of the disease state, the method comprising administering to a subject a first compound that converts to a second compound in vivo, wherein the second compound inhibits TRK activity in vivo. It is noted that the compounds of the present invention may be either pre-or post-conversion compounds.
In variations of each of the above methods, the disease state is selected from: cancerous proliferative diseases (e.g., brain, lung, squamous cell, bladder, stomach, pancreas, breast, head, neck, renal area (renal), kidney, ovary, prostate, colorectal, epidermal, esophageal, testicular, gynecological or thyroid cancer); non-cancerous proliferative diseases (e.g., benign skin hyperplasia (e.g., psoriasis), restenosis, and Benign Prostatic Hypertrophy (BPH)); pancreatitis; kidney disease; pain; preventing implantation of blastocysts; treating diseases associated with angiogenesis or vasculogenesis (e.g., tumor angiogenesis, acute and chronic inflammatory diseases such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, kaposi's sarcoma and ovarian cancer, breast cancer, lung cancer, pancreatic cancer, prostate cancer, colon cancer, and epidermoid cancer); asthma; neutrophil chemotaxis (e.g., reperfusion injury from myocardial infarction and stroke and inflammatory arthritis); septic shock; t cell mediated diseases where immunosuppression is valuable (e.g., prevention of organ transplant rejection, graft versus host disease, lupus erythematosus, multiple sclerosis, and rheumatoid arthritis); atherosclerosis; inhibiting keratinocytes responsive to the growth factor mixture; chronic Obstructive Pulmonary Disease (COPD) and other diseases.
In another aspect, the invention provides a method of treating a disease state for which a mutation in the TRK gene contributes to the pathology and/or symptomology of the disease state, such as melanoma, lung cancer, colon cancer and other types of tumors.
In another aspect, the present invention relates to the use of compounds and variants of any one of the above embodiments as medicaments. In another aspect, the present invention relates to the use of a compound of any one of the above embodiments and variants for the manufacture of a medicament for inhibiting TRK.
In another aspect, the present invention relates to the use of compounds and variants of any one of the above embodiments for the manufacture of a medicament for the treatment of pathological and/or symptomatic disease states caused by TRK activity.
Administration and pharmaceutical compositions
Generally, the compounds of the present invention will be administered in a therapeutically effective amount, either alone or in combination with one or more therapeutic agents, by any of the usual and acceptable means known in the art. The therapeutically effective amount may vary widely depending on the severity of the disease, age and relative health of the subject, the potency of the compound used and other factors known in the art. For example, for the treatment of neoplastic diseases and immune system diseases, the required dosage will vary depending upon the mode of administration, the particular condition being treated and the desired effect.
In general, satisfactory results are achieved at daily dosages of from 0.001 to 100mg/kg body weight, in particular from about 0.03 to 2.5mg/kg body weight. Daily doses for larger mammals, such as humans, may be administered in a convenient form, for example in divided doses up to four times a day or in sustained release form, from about 0.5mg to about 2000mg, or more specifically, from 0.5mg to 1000 mg. Suitable unit dosage forms for oral administration contain from about 1 to 50mg of the active ingredient.
The compounds of the present invention may be administered in the form of pharmaceutical compositions, by any conventional route; e.g., enterally, e.g., orally, e.g., in the form of tablets or capsules, parenterally, e.g., in the form of injectable solutions or suspensions; or topically, e.g., as a lotion, gel, ointment or cream, or in nasal or suppository form.
Pharmaceutical compositions containing a compound of the invention in free base or pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent may be manufactured in conventional manner by means of mixing, granulating, coating, dissolving or lyophilizing processes. For example, pharmaceutical compositions comprising a compound of the invention in combination with at least one pharmaceutically acceptable carrier or diluent may be formulated in conventional manner by admixture with a pharmaceutically acceptable carrier or diluent. Unit dosage forms for oral administration contain, for example, from about 0.1mg to about 500mg of active substance.
In one embodiment, the pharmaceutical composition is a solution, including a suspension or dispersion, such as an isotonic aqueous solution, of the active ingredient. In the case of lyophilized compositions comprising the active ingredient alone or in admixture with a carrier such as mannitol, dispersions or suspensions may be prepared prior to use. The pharmaceutical compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. Suitable preservatives include, but are not limited to, antioxidants such as ascorbic acid, microbicides such as sorbic acid or benzoic acid. The solution or suspension may also contain a thickening agent including, but not limited to, sodium carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatin, or a solubilizing agent such as tween 80 (polyoxyethylene (20) sorbitan monooleate).
Suspensions in oil may contain, as oily component, vegetable oils, synthetic or semisynthetic oils, commonly used for injection purposes. Examples include liquid fatty acid esters containing as the acid component a long chain fatty acid having from 8 to 22 carbon atoms, or in some embodiments, from 12 to 22 carbon atoms. Suitable liquid fatty acid esters include, but are not limited to, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, arachidic acid, behenic acid or corresponding unsaturated acids, such as oleic acid, elaidic acid, erucic acid, brassidic acid and linoleic acid, if desired, with antioxidants, such as vitamin E, 3-carotene or 3, 5-di-tert-butyl-hydroxytoluene. The alcohol component of these fatty acid esters may have six carbon atoms and may be monovalent or polyvalent, such as mono-, di-or trivalent alcohols. Suitable alcohol components include, but are not limited to, methanol, ethanol, propanol, butanol or pentanol or isomers thereof, ethylene glycol and glycerol.
Other suitable fatty acid esters include, but are not limited to, ethyl oleate, isopropyl myristate, isopropyl palmitate,
Figure BDA0003541883110000321
m2375, (polyoxyethylene glycerol),
Figure BDA0003541883110000322
m1944 CS (unsaturated polyglycolyzed glyceride prepared by alcoholysis of oleum Armeniacae amarum, containing glyceride and polyethylene glycol ester), LABRASOLTM(saturated PEGylated glycerides prepared by alcoholysis of TCM, containing glycerides and polyethylene glycol esters; all available from GaKefosse, France), and/or
Figure BDA0003541883110000323
812 (triglycerides of saturated fatty acids with a chain length of from C8 to C12, from huls AG, germany), and vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil or peanut oil.
Pharmaceutical compositions for oral administration may be obtained, for example, by mixing the active ingredient with one or more solid carriers, if desired granulating a resulting mixture, and processing the mixture or granules by adding further excipients, in the form of tablets or tablet cores.
Suitable carriers include, but are not limited to, fillers, for example sugars, such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and binders, for example starches, such as corn, wheat, rice or potato starch, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrants, such as the above-mentioned starches, carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate. Additional excipients include flow-regulating and lubricating agents, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol, or derivatives thereof.
The tablet cores may be provided with a suitable, optionally enteric, coating by using, inter alia, a concentrated sugar solution, which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or a coating solution in a suitable organic solvent or solvent mixture, or, for enteric coatings, a solution of a suitable cellulose preparation, such as an acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate solution. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes or to indicate different doses of the active ingredient.
Pharmaceutical compositions for oral administration may also include hard capsules, including gelatin or soft, sealed capsules containing gelatin and a plasticizer, such as glycerol or sorbitol. Hard capsules may contain the active ingredients in the form of granules, for example in admixture with fillers such as corn starch, binders and/or glidants such as talc or magnesium stearate, and optionally stabilizers. In soft capsules, the active ingredient may be dissolved or suspended in suitable liquid excipients, such as fatty oils, paraffin oil or liquid polyethylene glycols or fatty acid esters of ethylene glycol or propylene glycol, to which stabilizers and detergents, for example of the fatty acid ester type of polyoxyethylene sorbitol, may also be added.
Pharmaceutical compositions suitable for rectal administration, for example suppositories, comprise a combination of the active ingredient and a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.
Pharmaceutical compositions suitable for parenteral administration may contain the active ingredient in water-soluble form, for example as a water-soluble salt or as an aqueous injection suspension containing a viscosity-increasing substance, for example sodium carboxymethylcellulose, an aqueous solution of sorbitol and/or dextran, and, if desired, a stabilizer. The active ingredient, optionally together with excipients, may also be in a lyophilized form and may be prepared as a solution by addition of a suitable solvent prior to parenteral administration. The solutions used, for example, for parenteral administration, can also be used as infusion solutions. Injectable preparations are generally prepared under sterile conditions, and filled, for example, in ampoules or vials, and in sealed containers.
The invention also provides a pharmaceutical combination, e.g. a kit, comprising a) a compound disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one auxiliary agent. The kit may contain instructions for its use.
Combination therapy
The compounds or pharmaceutically acceptable salts described herein may be used alone or in combination with other therapeutic agents.
For example, the use of an adjuvant (adjuvant) may enhance the therapeutic effect of a compound of the invention (e.g., the therapeutic benefit of an adjuvant drug alone may be minimal, but in combination with another drug, may enhance the therapeutic benefit of a subject), or, for example, the therapeutic benefit of a subject may be enhanced by the combination of a compound of the invention with another therapeutic agent that is also therapeutically effective. For example, in the treatment of gout, the compound of the invention may be used in combination with another drug for gout therapy to enhance clinical benefit. Alternatively, for example, if the adverse effect of using the compounds of the present invention is nausea, then an anti-nausea agent may be used in combination. Alternatively, combination therapies may include, but are not limited to, physical therapy, psychotherapy, radiation therapy, compression therapy of diseased areas, rest, dietary improvement, and the like. Regardless of the disease, disorder, or condition, both therapies should have additive or synergistic effects to benefit the treatment of an individual.
Where the compounds of this patent are used in combination with other therapeutic agents, the pharmaceutical compositions of the compounds of this patent may be administered by the same route as the other drugs, or by different routes due to differences in physical and chemical properties. For example, oral administration of a compound of this patent may produce and maintain good blood levels, while intravenous administration of another therapeutic agent may be required. Thus, the compound of this patent and the other therapeutic agent may be administered simultaneously, sequentially or separately.
The compounds of formula (I) are expected to be effective in combination with one or more of the following: alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotics, antiproliferatives, antivirals, aurora kinase inhibitors, promoters of other apoptosis (e.g., Bcl-xL, BCL-w, and Bfl-1) inhibitors, death receptor pathway activators, Bcr-Abl kinase inhibitors, antibodies to BiTE (bispecific T-cell engagers), antibody drug conjugates, biological response modifiers, cyclin-dependent kinase inhibitors, cell cycle inhibitors, cyclooxygenase-2 ErbB inhibitors, DVDs, leukemia virus oncogene homolog (2) receptor inhibitors, growth factor inhibitors, Heat Shock Protein (HSP) -90 inhibitors, histone acetylases (HDAC) inhibitors, hormonal therapies, immunological agents, inhibitors of apoptosis protein Inhibitors (IAPs), intercalating antibiotics, inhibitors of apoptosis proteins, Kinase inhibitors, kinesin inhibitors, JAK2 inhibitors, rapamycin inhibitors for mammals, micrornas, mitogen-activated extracellular signal-regulated kinase inhibitors, multivalent binding proteins, non-steroidal anti-inflammatory drugs (NSAIDs), poly ADP (adenosine diphosphate) -ribose polymerase (PARP) inhibitors, platinum-based chemotherapeutic drugs, polo-like kinase (Plk) inhibitors, phosphoinositide 3 kinase (PI3K) inhibitors, proteasome inhibitors, purine analogs, pyrimidine analogs, receptor tyrosine kinase inhibitors, retinoid/deltoid plant alkaloids, small interfering RNAs (sirnas), topoisomerase inhibitors, ubiquitin ligase inhibitors, and the like.
Examples
There are various methods for synthesizing the compound of formula (I) or a pharmaceutically acceptable salt thereof, and representative methods are listed in this example. However, it is to be noted that the compounds of formula (I) or pharmaceutically acceptable salts thereof may also be obtained by synthesis in other synthetic schemes.
In certain compounds of formula (I), the attachment of atoms to other atoms may result in the presence of particular stereoisomers (e.g. chiral centres). The synthesis of a compound of formula (I) or a pharmaceutically acceptable salt thereof may result in a mixture of different isomers (enantiomers, diastereomers). Unless a particular configuration is specified, all recited compounds include different stereoisomers that may exist.
The compounds of formula (I) may also be prepared as pharmaceutically acceptable acid addition salts, for example by reacting the free base form of the compounds of the invention with a pharmaceutically acceptable inorganic or organic acid. Or a compound of formula (I) in free acid form with a pharmaceutically acceptable inorganic or organic base, to form a pharmaceutically acceptable base addition salt. Inorganic and organic acids and bases suitable for the preparation of pharmaceutically acceptable salts of the compounds of formula (I) are described in the definitions section of this application. In addition, salt forms of the compounds of formula (I) may also be prepared by using salts of the starting materials or intermediates.
The free acid or free base of the compound of formula (I) may be prepared from the corresponding base addition salt or acid addition salt thereof. The acid addition salt forms of the compounds of formula (I) may be converted to the corresponding free base, for example by treatment with a suitable base such as ammonium hydroxide solution, sodium hydroxide and the like. Base addition salt forms of the compounds of formula (I) may be converted to the corresponding free acids, for example by treatment with a suitable acid such as hydrochloric acid and the like.
An N-oxide of a compound of formula (I) or a pharmaceutically acceptable salt thereof may be prepared by methods known in the art. For example, the N-oxide can be obtained by reacting a non-oxidized form of the compound of formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, peroxymaleic acid (permaleic acid), perbenzoic acid, peracetic acid, m-chloroperoxybenzoic acid, etc.) in an inert organic solvent (e.g., halogenated hydrocarbon such as dichloromethane) at 0 to 80 ℃. Alternatively, the N-oxides of the compounds of formula (I) may also be prepared from the N-oxides of the starting materials.
The non-oxidized compound of formula (I) can be prepared by reacting N-oxide thereof with a reducing agent (such as sulfur, sulfur dioxide, triphenylphosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, phosphorus tribromide and the like) in a corresponding inert organic solvent (such as acetonitrile, ethanol, dioxane aqueous solution and the like) at 0-80 ℃.
Protected derivatives of compounds of formula (I) may be prepared by methods well known to those skilled in the art. For a detailed technical description of the addition and removal of protecting groups see: greene, Protecting Groups in Organic Synthesis,3rd edition, John Wiley & Sons, Inc.1999.
The methods, routes and labels and general knowledge used in the examples are in accordance with the current scientific literature, e.g., the journal of the American chemical Association or the journal of biochemistry. Unless otherwise indicated, standard single or three letter abbreviations generally refer to L-amino acid residues. All starting materials used were purchased from commercial suppliers and used without further purification unless otherwise indicated. For example, the following abbreviations are used in the examples and throughout the specification: g (g), mg (mg), L (L), mL (mL), μ L (μ L), psi (pounds per square inch), M (mol), mM (mmol), i.v. (i.v.), Hz (Hz), MHz (megahertz), mol (mol), mmol (mmol), RT (ambient temperature), min (min), h (h), mp (melting point), TLC (thin layer chromatography), RT (retention time), RP (reversed phase), MeOH (methanol), i-PrOH (isopropanol), TEA (triethylamine), TFA (trifluoroacetic acid), TFAA (trifluoroacetic anhydride), THF (tetrahydrofuran), DMSO (dimethyl sulfoxide), EtOAc (ethyl acetate), DME (1, 2-dimethylethane), DCM (dichloromethane), DCE (dichloroethane), DMF (N, N-dimethylformamide), DMPU (N, N' -dimethylpropyleneurea), CDI (1, 1-carbonyldiimidazole), IBCF (isobutyl chloroformate), HOAc (acetic acid), HOSu (N-hydroxysuccinimide), HOBT (1-hydroxybenzotriazole), Et2O (diethyl ether), EDCI (1- (3-dimethylaminopropyl) 3-ethylcarbodiimide hydrochloride), BOC (t-butyloxycarbonyl), FMOC (9-fluorenylmethoxycarbonyl), DCC (dicyclohexylcarbodiimide), CBZ (benzyloxycarbonyl), Ac (acetyl), atm (atmospheric pressure), TMSE (2- (trimethylsilyl) ethyl), TMS (trimethylsilyl), TIPS (triisopropylsilyl), TBS (t-butyldimethylsilyl), DMAP (4-dimethylaminopyridine), Me (methyl), OMe (methoxy), Et (ethyl), tBu (t-butyl), HPLC (high Performance liquid chromatography), BOP (2-oxobis (2-oxoethyl) and the like-3-oxazolidinyl) hypophosphoryl chloride), TBAF (tetra-n-butylammonium fluoride), mCPBA (m-chloroperoxybenzoic acid).
Ether or Et2O is all ether; brine is then a saturated aqueous NaCl solution. Unless otherwise indicated, all temperatures refer to degrees Celsius (degrees Celsius) and all reactions are carried out in an inert atmosphere at room temperature.
1H NMR spectra were recorded using a Varian Mercury Plus 400 NMR spectrometer. Chemical shifts are expressed in ppm. The coupling constants are all in hertz (Hz). Apparent diversity is described in the split mode and is assigned as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet) and br (broad).
Low resolution Mass Spectrometry (MS) and compound purity data were from a Shimadzu LC/MS single quadrupole system equipped with an electrospray ion detector (ESI), ultraviolet detectors (220 and 254nm) and an Evaporative Light Scattering Detector (ELSD). Thin layer chromatography was performed using 0.25mm Asahi-poise silica gel plate (60F-254), 5% ethanol phosphomolybdate solution, ninhydrin or p-methoxybenzaldehyde solution and observing under an ultraviolet lamp. Silica gel (200-300 mesh, Qingdao ocean chemical Co., Ltd.) is used for the rapid column chromatography.
Synthetic schemes
The synthetic schemes for all compounds of the present invention are illustrated by the following schemes and examples. The starting materials used are commercially available or may be prepared according to established procedures or by methods exemplified herein.
The intermediates listed in the following figures are obtained from the literature or synthesized according to methods well known to those skilled in the art.
As an illustration, scheme 1 discloses a method of synthesis of the presently disclosed compounds of formula I. Intermediate IV is prepared by nucleophilic substitution of heteroaryl II with fused heterocycle III as shown in the synthetic scheme. Hydrolysis of ester IV produces carboxylic acid V. V is further transformed to obtain the compound of the formula I.
Figure BDA0003541883110000361
As an example of the preparation of the compound of formula III, one synthetic route for the compound of formula IIIa is shown in FIG. 2. Starting from IIIa-A, either commercially available or known in the literature. The lactones IIIa-B can be prepared by reacting (S) -2- (chloromethyl) oxirane with IIIa-A. The IIIa-B is hydrolyzed to produce compounds of formula IIIa-C which are further converted to IIIa-D. Oxidation of IIIa-D affords the aldehydes IIIa-E. Intermediates IIIa-H can be prepared from aldehydes IIIa-E by reaction with Wittig reagents, followed by hydroboration-oxidation of the double bond and hydrolysis of the ester. Protection of the free hydroxyl group of IIIa-H affords compounds of formula IIIa-I, which are further converted to IIIa-J by Curtius rearrangement. The intermediates IIIa-M can be obtained by three steps of deprotection of the protecting groups of IIIa-J, methanesulfonic acid of the hydroxyl groups of IIIa-L, and intramolecular cyclization in the presence of a base. Finally, deprotection of Cbz in IIIa-M affords compounds of formula IIIa.
Figure BDA0003541883110000371
By way of illustration, scheme 3 discloses a method of synthesis of the presently disclosed compounds of formula I. As shown in the scheme, decarboxylation of intermediate carboxylic acid V affords the intermediate of formula VI. Halogenation of compound VI with N-iodosuccinimide yields intermediate VII, which upon transition metal catalyzed cross-coupling reaction yields the compound of formula I.
Figure BDA0003541883110000372
By way of further illustration, scheme 4 discloses a method of synthesis of the presently disclosed compounds of formula I. As shown in the scheme, intermediate boronic acid VIII is prepared by exchange of intermediate metal halide of formula VII followed by quenching with trimethyl borate or triisopropoxyborane. Coupling reaction is carried out on the boric acid VIII under the catalysis of palladium to obtain the compound shown in the formula I.
Figure BDA0003541883110000373
In some cases, the above synthetic schemes may be ordered as appropriate in order to facilitate the reaction or to avoid the production of unnecessary reaction products. In order that the invention may be more fully understood, the following examples are set forth. These examples are only examples and should not be construed as limiting the invention.
Example 1
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-amines (1)
Figure BDA0003541883110000381
(1R,5S) -1- (2, 5-difluorophenyl) -3-oxabicyclo [3.1.0]Hexane-2-one (1a)
To a solution of 2- (2, 5-difluorophenyl) acetonitrile (5.00g,32.7mmol) and (S) -2- (chloromethyl) oxirane (4.50g,49.0mmol) in THF (40mL) at-20 deg.C was added NaHMDS (42.0mL,81.8mmol) dropwise. The mixture was stirred at-15 ℃ for 3 h. The reaction was quenched with water and concentrated. Ethanol (30mL) and potassium hydroxide (5.50g, 38.2mmol) were added to the mixture and stirred at 80 ℃ overnight. The mixture was adjusted to pH 2-3 with concentrated HCl and then stirred at 60 ℃ for 2 hours. The mixture was extracted with EtOAc. With saturated NaHCO3The organic phase is washed with an aqueous solution, Na2SO4Drying and concentrating. The residue was purified by column chromatography on silica gel to give the title compound (1R,5S) -1- (2, 5-difluorophenyl) -3-oxabicyclo [3.1.0]Hexane-2-one (1 a). MS-ESI (M/z) 211[ M + 1]]+
(1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) cyclopropane-1-carboxylic acid (1b)
To (1R,5S) -1- (2, 5-difluorophenyl) -3-oxabicyclo [3.1.0] at room temperature]Hexane-2-one (1a) (4.20g,20.0mmol) in MeOH/THF/H2To the O (16/16/32mL) solution was added LiOH2O (4.20g,100 mmol). Stirred at room temperature for 3 hoursAfter this time, the reaction was quenched with water and adjusted to pH 4-5 with 6N HCl. The mixture was extracted with EtOAc (4X 80 mL). The extract was washed with brine (100mL), Na2SO4Dried and concentrated to give crude (1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) cyclopropane-1-carboxylic acid (1b) which was used in the next step without further purification. MS-ESI (M/z):229[ M +1 [)]+
Ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) cyclopropane-1-carboxylate (1c)
Mixing (1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) cyclopropane-1-carboxylic acid (1b) (100mg,0.439mmol), KHCO3A mixture of (57.0mg,0.570mmol) and EtBr (144mg,1.32mmol) in DMF (5mL) was stirred at 23 ℃ for 4 h. The reaction was quenched with water and extracted with ethyl acetate. The extract was washed with brine (30mL), Na2SO4Drying and concentration gave crude ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) cyclopropane-1-carboxylate (1c) which was used in the next step without further purification. MS-ESI (M/z):257[ M + 1]]+
Ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2-formylcyclopropane-1-carboxylate (1d)
To a solution of ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2- (hydroxymethyl) -cyclopropane-1-carboxylate (1c) (98.0mg,0.380mmol) in DCM (4mL) at rt was added DMP (243mg,0.570 mmol). The mixture was stirred at room temperature for 4 hours and saturated NaHCO was used3And (4) quenching the aqueous solution. The mixture was extracted with DCM. The extract was washed with brine, Na2SO4Dried and the solvent concentrated. The residue was purified by column chromatography on silica eluting with PE/EtOAc (10:1) to give the title compound ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2-formylcyclopropane-1-carboxylate (1 d). MS-ESI (M/z):255[ M + 1[ ]]+
Ethyl (1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylate (1e)
To a solution of ethyl (1R,2S) -1- (2, 5-difluorophenyl) -2-formylcyclopropane-1-carboxylate (1d) (100mg,0.394mmol) and methyltriphenylphosphonium iodide (207mg,0.512mmol) in DCM (3mL) at 0 deg.C was added t-BuOK (62.0mg,0.552 mmol). The mixture was stirred at 0 ℃ for 0.5 h. The mixture was concentrated to give a crude product of ethyl (1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylate (1e), which was used directly in the next step.
(1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylic acid (1f)
Ethyl (1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylate (1e) (99.3mg,0.394mmol) and NaOH (158mg,3.94mmol) in MeOH/H2The mixture in O (1/1.5mL) was stirred at 55 ℃ for 4 h. The reaction was quenched with water and washed with DCM. The aqueous layer was acidified with hydrochloric acid to pH 3-4. The mixture was extracted with ethyl acetate. The extract was washed with brine (30ml), Na2SO4Dried and the solvent concentrated. The residue was purified by column chromatography on silica eluting with PE/EtOAc (5:1) to give the title compound (1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylic acid (1 f). MS-ESI (M/z) 225[ M + 1]]+
(1R,2R) -1- (2, 5-difluorophenyl) -2- (2-hydroxyethyl) cyclopropane-1-carboxylic acid (1g)
To a solution of (1R,2R) -1- (2, 5-difluorophenyl) -2-vinylcyclopropane-1-carboxylic acid (1f) (30.0mg,0.134mmol) in THF (1mL) at 0 deg.C was added BH dropwise3Of THF (0.33mL,0.33 mmol). The mixture was stirred at room temperature for 0.2 h. NaOH (6N, 0.2mL) and H were then added to the mixture at room temperature2O2(30%, 152mg, 1.34mmol) and stirred at room temperature for 20 min. The reaction was quenched with water and washed with DCM. The aqueous layer is acidified with hydrochloric acid to a pH of 3-4. The mixture was extracted with ethyl acetate. Washed with brine (100mL), Na2SO4Drying and concentration of the solvent gave the crude product (1R,2R) -1- (2, 5-difluorophenyl) -2- (2-hydroxyethyl) cyclopropane-1-carboxylic acid (1g) which was used directly in the next reaction. MS-ESI (M/z):243[ M + 1[ ]]+
(1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropane-1- Formic acid (1h)
At 0 deg.C, adding into (1R,2R) -1-, (To a solution of 2, 5-difluorophenyl) -2- (2-hydroxyethyl) cyclopropane-1-carboxylic acid (1g) (385mg,1.60mmol) in DCM/DMF (4mL/2mL) were added TBSCl (483mg,3.20mmol) and imidazole (433mg,6.40 mmol). The mixture was stirred at 20 ℃ overnight and concentrated. Saturated Na2CO3The mixture was diluted with aqueous solution (30mL) and washed with ethyl acetate. The aqueous layer was acidified to pH 3-4 with hydrochloric acid and extracted with ethyl acetate. Brine (100mL), Na2SO4Drying and concentration gave the crude product (1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropane-1-carboxylic acid (1h) which was used directly in the next reaction. MS-ESI (M/z):357[ M + 1[ ]]+
Benzyl ((1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropyl Yl) carbamic acid ester (1i)
To a solution of (1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropane-1-carboxylic acid (1h) (383mg,1.08mmol) and BnOH (1.17g,10.8mmol) in toluene (8mL) at room temperature was added DPPA (446mg,1.62mmol) and TEA (273mg,2.70 mmol). The mixture was stirred at 85 ℃ overnight. The reaction was quenched with water and extracted with ethyl acetate. The extract was washed with brine (30ml), Na2SO4Dried and the solvent concentrated. The residue was purified by column chromatography on silica gel eluting with PE/EtOAc (20:1) to give the title compound benzyl ((1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropyl) carbamate (1 i). MS-ESI (M/z):462[ M + 1[ ]]+
Benzyl ((1R,2R) -1- (2, 5-difluorophenyl) -2- (2-hydroxyethyl) cyclopropyl) carbamate (1j)
To a solution of benzyl ((1R,2R) -2- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1- (2, 5-difluorophenyl) cyclopropyl) carbamate (1i) (15.0mg,0.033mmol) in THF (1mL) at room temperature was added TBAF (1M,0.16 mL). The mixture was stirred at 25 ℃ for 1 hour. The reaction was quenched with water and extracted with EtOAc. The extract was washed with brine (30ml), Na2SO4Drying and concentrating to obtain crude benzyl ((1R,2R) -1- (2, 5-difluorophenyl)) The (2-hydroxyethyl) cyclopropyl) carbamate (1j) was used directly in the next reaction. MS-ESI (M/z):348[ M + 1]]+
2- ((1R,2R) -2- (((benzyloxy) carbonyl) amino) -2- (2, 5-difluorophenyl) cyclopropyl) ethylmethanesulfonic acid Ester (1k)
To a solution of benzyl ((1R,2R) -1- (2, 5-difluorophenyl) -2- (2-hydroxyethyl) cyclopropyl) carbamate (1j) (246mg,0.710mmol) and MsCl (122mg,1.06mmol) in DCM (2mL) at 0 deg.C was added TEA (180mg,1.76 mmol). The mixture was stirred at 0 ℃ for 30 minutes. The reaction was quenched with water and the mixture was extracted with DCM. Washing the extract with 1N HCl, water and brine in sequence, Na2SO4Drying and concentration gave the crude product 2- ((1R,2R) -2- (((benzyloxy) carbonyl) amino) -2- (2, 5-difluorophenyl) -cyclopropyl) ethyl methanesulfonate (1k) which was used directly in the next reaction. MS-ESI (M/z) 426[ M + 1]]+
Benzyl (1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-carboxylate (1l)
To a solution of 2- ((1R,2R) -2- (((benzyloxy) carbonyl) amino) -2- (2, 5-difluorophenyl) cyclopropyl) ethyl methanesulfonate (1k) (302mg,0.710mmol) in DMF (6mL) at 0 ℃ was added NaH (60%, 43 mg). The mixture was stirred at 25 ℃ for 0.5 h. The reaction was quenched with water. The mixture was extracted with ethyl acetate and concentrated. The residue was purified by column chromatography on silica eluting with PE/EtOAc (15: 1-10: 1) to give the title compound benzyl (1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]]Hexane-2-carboxylate (1 l). MS-ESI (M/z) 330[ M + 1]]+
(1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane (1m)
Benzyl (1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-carboxylate (1l) (40.0mg,0.122mmol) and conc.HCl (1mL) in MeOH/CH3The mixture in CN (0.1mL/0.5mL) was stirred at 60 ℃ overnight. The mixture was basified with aq. naoh to pH 10 and the aqueous phase was extracted with DCM. The extract was washed with brine (30ml), Na2SO4Drying and concentrating to obtain crude product (1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane (1m) was used directly in the next reaction. MS-ESI (M/z) 196[ M + 1]]+
Ethyl 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hex-2-yl) -pyrazolo [1, 5-a]pyrimidine-3-carboxylic acid ester (1n)
To (1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] at room temperature]Hexane (1m) (27.0mg,0.138mmol) and TEA (35.0mg,0.345mmol) in DMF (1.5mL) was added ethyl 5-chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylate (36.0mg,0.160 mmol). The mixture was stirred at 50 ℃ for 1.5h under nitrogen. The reaction was quenched with water and the mixture was extracted with ethyl acetate. The extract was washed with brine (30ml), Na2SO4Dried and concentrated. The residue was purified by column chromatography on silica eluting with PE/EtOAc (1.5:1) to give the title compound ethyl 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]-Hexane-2-yl) -pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ester (1 n). MS-ESI (M/z):358[ M + 1]]+
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a] Pyrimidine-3-carboxylic acid (1o)
Ethyl 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]-Hexane-2-yl) -pyrazolo [1,5-a]Pyrimidine-3-carboxylate (1n) (1.22g,3.18mmol) and lioh2O (763mg,31.8mmol) in MeOH/H2The mixture in O (30mL/3mL) was stirred at 70 ℃ overnight. The reaction mixture was diluted with water and acidified with 6N HCl to pH 3-4. The mixture was extracted with DCM. Washing the extract with brine, Na2SO4Drying and concentrating to give 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (1o), was used directly in the next reaction. MS-ESI (M/z):357[ M +1 [)]+
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- a]Pyrimidine-3-carbonyl) hydrazine-1-thioamide (1p)
Mixing 5- ((1R,5S) -1- (2)5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (1o) (500mg,1.40mmol), HATU (585mg,1.54mmol) and Et3A mixture of N (300. mu.L, 2.10mmol) in DMF (6mL) was stirred at room temperature for 1.5 h. Thiosemicarbazide (141mg, 1.54mmol) was then added and the mixture stirred at room temperature overnight. The mixture was concentrated, then water (20mL) was added. After stirring for 1 hour, the mixture was filtered. H for filter cake2O wash to give the title compound as crude 2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carbonyl) hydrazine-1-thioamide (1 p). MS-ESI (M/z) 430[ M + 1]]+
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-amines (1)
PPA (27g) was preheated to 100 ℃ and 2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] was added to the hot PPA solution at 100 ℃]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carbonyl) hydrazine-1-thioamide (1p) (602mg,1.40 mmol). The mixture was stirred at 100 ℃ for 2 h. The reaction mixture was cooled to room temperature and poured into ice, over NH3.H2And O, adjusting the pH of the mixture to 9-10. Filtration and dissolution of the residue in DCM and drying (Na)2SO4) And concentrated to give 5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-amine (1). MS-ESI (M/z) 412[ M + 1]]+
Example 2
4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl morpholine (2)
Figure BDA0003541883110000411
2-bromo-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) Pyrazolo of the formula [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazole (2a)
To 5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] at room temperature]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-amine (1) (41.1mg,0.10mmol) and CuBr2(27.0mg,0.12mmol) of CH3CN (2mL) mixture was added t-BuONO (12.4mg,0.12 mmol). After addition, the mixture was heated to 65 ℃ and stirred at 65 ℃ for 2 hours. The reaction was cooled to RT, quenched with water and extracted with EtOAc. Washing the extract with water and brine, Na2SO4Drying and concentrating to obtain 2-bromo-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazole (2a) crude product was used in the next step without further purification. MS-ESI (M/z) 475,477[ M + 1]]+
4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl morpholine (2)
Reacting 2-bromo-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]]Hexane-2-yl) pyrazolo [1,5-a]A mixture of pyrimidin-3-yl) -1,3, 4-thiadiazole (2a) (23mg,0.049mmol), morpholine (6.3mg,0.072mmol) and DIPEA (13mg,0.098mmol) in DMF (1mL) was stirred at 120 ℃ overnight. The reaction was cooled to rt, concentrated and extracted with EtOAc. The extract was washed with water and brine, Na2SO4Drying and concentrating. The residue was purified by PTLC eluting with DCM/MeOH (25:1) to give the title compound 4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) morpholine (2). MS-ESI (M/z):482[ M + 1]]+
Example 3
Methyl (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) carbamates (3)
Figure BDA0003541883110000421
To 5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] at 0 deg.C]Hexane-2-yl) pyrazolo [1,5-a]To a solution of pyrimidin-3-yl) -1,3, 4-thiadiazol-2-amine (1) (20mg,0.050mmol) and DIPEA (8.4mg,0.065mmol) in DCM (1mL) was added methyl chloroformate (5.2mg,0.055 mmol). The mixture was stirred at room temperature overnight. The mixture was extracted with DCM. Washing the extract with brine, Na2SO4Dried and then concentrated. The residue was purified by PTLC eluting with DCM/MeOH (25:1) to give the title compound methyl (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) carbamate (3). MS-ESI (M/z):470[ M + 1[ ]]+
Example 4
2- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) propan-2-ol (4)
Figure BDA0003541883110000431
5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -N' - (2-hydroxy-2-methyl Propionyl) pyrazolo [1,5-a]Pyrimidine-3-carbohydrazide (4a)
To 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxylic acid (1o) (50mg,0.14mmol) and 2-hydroxy-2-methylpropanehydrazide (50mg,0.42mmol) in DMF (1mL) was added HATU (106mg,0.28mmol) and DIPEA (54mg,0.42 mmol). The mixture was stirred at room temperature overnight. The mixture was diluted with ethyl acetate. Water and brine wash the extract, Na2SO4Drying and concentrating to obtain 5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -N' - (2-hydroxy-2-methylpropionyl) pyrazolo [1,5-a]Pyrimidine-3-carbohydrazide (4 a). MS-ESI (M/z) 457[ M +1]+
N' - (2- ((tert-butyldiphenylsilyl) oxy) -2-methylpropionyl) -5- ((1R) -1- (2, 5-difluorobenzene Yl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carbohydrazide (4b)
At room temperature to 5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -N' - (2-hydroxy-2-methylpropionyl) pyrazolo [1,5-a]To a solution of pyrimidine-3-carbohydrazide (4a) (64mg,0.14mmol) and imidazole (15mg,0.21mmol) in DMF (1mL) was added TBDPSCl (46mg,0.17 mmol). The mixture was stirred at 50 ℃ overnight. The mixture was diluted with ethyl acetate. Water and brine wash the extract, Na2SO4Dried and concentrated. The residue was purified by PTLC eluting with DCM/MeOH (20:1) to give the title compound N' - (2- ((tert-butyldiphenylsilyl) oxy) -2-methylpropanoyl) -5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidine-3-carbohydrazide (4 b). MS-ESI (M/z):695[ M + 1]]+
2- (2- ((tert-butyldiphenylsilyl) oxy) propan-2-yl) -5- (5- ((1R) -1- (2, 5-difluorophenyl) - 2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazole (4c)
Reacting N' - (2- ((tert-butyldiphenylsilyl) oxy) -2-methylpropanoyl) -5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]A mixture of pyrimidine-3-carbohydrazide (4b) (13mg,0.019mmol) and Lawson's reagent (15mg,0.037mmol) in toluene (1mL) was stirred at 65 ℃ for 3 h. The mixture was cooled to room temperature and concentrated. The residue was purified by column chromatography on silica gel eluting with DCM/MeOH (20:1) to give 2- (2- ((tert-butyldiphenylsilyl) oxy) propan-2-yl) -5- (5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazole (4 c). MS-ESI (M/z):693[ M + 1]]+
2- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) propan-2-ol (4)
2- (2- ((tert-butyl diphenyl silicon) oxy) propane-2-yl) -5- (5- ((1R) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]A mixture of pyrimidin-3-yl) -1,3, 4-thiadiazole (4c) (11mg,0.016mmol) in TBAF (1M in THF, 1mL) was stirred at room temperature for 3 h. The mixture was diluted with ethyl acetate. Water and brine wash the extract, Na2SO4Dried and concentrated. The residue was purified by PTLC eluting with DCM/MeOH (20:1) to give the title compound 2- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) -1,3, 4-thiadiazol-2-yl) propan-2-ol (4). MS-ESI (M/z):455[ M +1 [)]+
Example 5
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- ((4-methyl) Piperazin-1-yl) methyl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (5)
Figure BDA0003541883110000441
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a] Pyrimidine (5a)
Mixing 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]A mixture of pyrimidine-3-carboxylic acid (1o) (310mg,0.87mmol) in 6N HCl (20mL) was stirred at 90 deg.C overnight. The mixture was cooled to room temperature and adjusted to pH 9-10 using NaOH, extracted with EtOAc. The extract was washed with water and brine in sequence, and Na2SO4Dried and concentrated to give the title compound 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]Crude product of pyrimidine (5 a). MS-ESI (M/z):313[ M + 1[ ]]+
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3-iodopyrazolo [1, 5-a]pyrimidine (5b)
Mixing 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) pyrazolo [1,5-a]A mixture of pyrimidine (5a) (129mg,0.410mmol) and NIS (102mg,0.451mmol) in DMF (2mL) was stirred at room temperature for 1.5 h. The mixture was cooled to 0 ℃ and Na2S2O3The aqueous solution was quenched and extracted with EtOAc. The extract was washed with water and brine in sequence, and Na2SO4Dried and concentrated to give the title compound 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3-iodopyrazolo [1,5-a]Crude product of pyrimidine (5 b). MS-ESI (M/z) 439[ M + 1]]+
(5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a] Pyrimidin-3-yl) boronic acid (5c)
To 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] at-78 deg.C]Hexane-2-yl) -3-iodopyrazolo [1,5-a]To a solution of pyrimidine (5b) (51.5mg,0.120mmol) in ultra dry THF (1.5mL) was added isopropyl magnesium chloride (2M in THF, 120. mu.L, 0.240mmol) dropwise, and the resulting mixture was stirred at-30 ℃ to-40 ℃ for 30 minutes. The reaction mixture was warmed to-25 ℃ and B (OMe) was added to the mixture3And (3) solution. After addition, the mixture was slowly warmed to room temperature and stirred at room temperature for 1.5 h. The mixture was cooled to 0 ℃ NH4Aqueous Cl solution quenched and EtOAc extraction. The extract was washed with brine, Na2SO4Dried and concentrated to give the title compound (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Crude product of pyrimidin-3-yl) boronic acid (5 c). MS-ESI (M/z):357[ M +1 [)]+
1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine (5d)
To a solution of 2-bromo-5-formylpyridine (310mg,2.69mmol) and 1-methylpiperazine (500mg,2.69mmol) in DCM (20mL) at room temperature was added NaBH (OAc)3(860mg,4.06mmol) and stirred overnight. With Na2CO3The reaction was quenched with aqueous solution and the mixture was extracted with DCM. Washed with brine, Na2SO4Drying and concentrating. Passing the residue through a silica gel columnPurification by chromatography eluting with DCM/MeOH (100: 1-30: 1) afforded the title compound 1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine (5 d). MS-ESI (M/z) 270,272[ M + 1]]+
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- ((4-methyl) Piperazin-1-yl) methyl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (5)
1- ((6-Bromopyridin-3-yl) methyl) -4-methylpiperazine (5d) (41mg,0.15mmol), (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) boronic acid (5c) (36mg,0.10mmol), PdCl2(dppf)2(7.5mg,0.010mmol) and K3PO4A mixture of (2M,0.2mL,0.4mmol) in 1, 4-dioxane (3mL) was stirred at 95 ℃ for 5 h. The mixture was cooled to rt and extracted with EtOAc. Water and brine wash the extract, Na2SO4Drying and concentrating. The residue was purified by PTLC eluting with DCM/MeOH (15:1) to give the title compound 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]]Hexane-2-yl) -3- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrazolo [1,5-a]A pyrimidine (5). MS-ESI (M/z):502[ M +1 [)]+
Example 6
5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl-3- (5- (4-methylpiper) Oxazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6)
Figure BDA0003541883110000451
Tert-butyl 4- (6-bromopyridin-3-yl) piperazine-1-carboxylate (6a)
2-bromo-5-iodopyridine (1.70g,6.0mmol), tert-butylpiperazine-1-carboxylate (0.97g,5.0mmol), Pd2(dba)3(230mg,0.25mmol), Xantphos (434mg,0.75mmol) and NaOtA mixture of Bu (1.40g,15mmol) in toluene was stirred at 90 ℃ for 3 h. The mixture was cooled to room temperature and washed with H2Dilution with O and EtOAc extractionTaking, washing the extract with brine, Na2SO4And (5) drying. Filtered and concentrated. The residue was purified by column chromatography on silica gel eluting with PE/EA (10:1 to 8:1) to give the title compound tert-butyl 4- (6-bromopyridin-3-yl) piperazine-1-carboxylate (6 a). MS-ESI (M/z):342[ M +1]+
Tert-butyl 4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazoles And [1,5-a ]]Pyrimidin-3-yl) pyridin-3-yl) piperazine-1-carboxylic acid ester (6b)
The title compound tert-butyl 4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) piperazine-1-carboxylate (6b) was prepared according to the synthetic method of 5 replacing 1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine with tert-butyl 4- (6-bromopyridin-3-yl) piperazine-1-carboxylate (6 a). MS-ESI (M/z):574[ M +1 [)]+
5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (piperazin-1-yl) Pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6c)
To tert-butyl 4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] at room temperature]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) piperazine-1-carboxylic acid ester (6b) in DCM (1mL) was added HCl/EtOAc (4M,3mL) and stirred at room temperature for 0.5 h. The mixture was concentrated and washed with H2Dilution with O, NaHCO3The mixture was adjusted to pH 8. Extracted with DCM/MeOH (10:1), Na2SO4Drying and concentration gave the title compound 5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (piperazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6 c). MS-ESI (M/z):474[ M +1]+
5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (4-methylpiperazine) Oxazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6)
The title compound 5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (4-methylpiperazin-1-yl) pirPyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6) was synthesized according to 5d replacing 2-bromo-5-formylpyridine and 1-methylpiperazine with 5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0%]Hexane-2-yl) -3- (5- (piperazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (6c) and HCHO (33% H)2O solution). MS-ESI (M/z):488[ M + 1]]+
Example 7
1- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- a]Pyrimidin-3-yl) pyridin-3-yl) piperidin-4-ol (7)
Figure BDA0003541883110000461
8- (6-Bromopyridin-3-yl) -1, 4-dioxa-8-azaspiro [4.5]Decane (7a)
2-bromo-5-iodopyridine (852mg,3.0mmol), 1, 4-dioxa-8-azaspiro [4.5 ]]Decane (644mg,4.5mmol), Pd2(dba)3(137mg,0.15mmol), Xantphos (260mg,0.45mmol) and NaOtA mixture of Bu (720mg,7.5mmol) in toluene was stirred at 80 ℃ for 2 h. The mixture was cooled to room temperature and the residue was purified by silica gel column chromatography eluting with PE/EA (20: 1-5: 1) to give the title compound 8- (6-bromopyridin-3-yl) -1, 4-dioxa-8-azaspiro [4.5 ]]Decane (7 a). MS-ESI (M/z) 299,301[ M + 1]]+
1- (6-Bromopyridin-3-yl) piperidin-4-one (7b)
To 8- (6-bromopyridin-3-yl) -1, 4-dioxa-8-azaspiro [4.5]To a solution of decane (7a) (250mg,0.84mmol) in dioxane (2mL) was added HCl (3N,3mL) and stirred at room temperature for 1 h. By H2The mixture was diluted with O, adjusted to PH 9 with NaOH (20% aq), then extracted with DCM, Na2SO4Drying and concentration gave the title compound 1- (6-bromopyridin-3-yl) piperidin-4-one (7 b). MS-ESI (M/z) 255,257[ M + 1]]+
1- (6-Bromopyridin-3-yl) piperidin-4-ol (7c)
To a solution of 1- (6-bromopyridin-3-yl) piperidin-4-one (7b) (212mg,0.840mmol) in THF/MeOH (5mL/0.1mL) at room temperature was added NaBH4(64.0mg,1.67mmol) and the resulting mixture was stirred at room temperature for 1 hour. By H2The mixture was diluted O (10mL), extracted with DCM (3X 10mL), Na2SO4Dried and concentrated to give the title compound 1- (6-bromopyridin-3-yl) piperidin-4-ol (7 c). MS-ESI (M/z):257,259[ M + 1]]+
1- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- a]Pyrimidin-3-yl) pyridin-3-yl) piperidin-4-ol (7)
The title compound 1- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) piperidin-4-ol (7) was synthesized according to the method of 5, replacing 1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine (5d) with 1- (6-bromopyridin-3-yl) piperidin-4-ol (7 c). MS-ESI (M/z):489[ M + 1]]+
Example 8
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (4- (oxa) hexane Cyclobutan-3-yl) piperazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (8)
Figure BDA0003541883110000471
1- (6-bromopyridin-3-yl) piperazine (8a)
To a solution of tert-butyl 4- (6-bromopyridin-3-yl) piperazine-1-carboxylate (6a) (0.50g,1.5mmol) in EtOAc (2mL) at room temperature was added HCl/EtOAc (4N,5mL) and the resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated and washed with H2Dilution with O and saturated Na2CO3The aqueous layer was adjusted to pH 10 and extracted with DCM/MeOH (10: 1). The extract was washed with brine, Na2SO4Dried and concentrated to give the title compound 1- (6-bromopyridin-3-yl) piperazine (8 a). MS-ESI (M/z) 242,244[ M [ ]+1]+
1- (6-bromopyridin-3-yl) -4- (oxetan-3-yl) piperazine (8b)
A mixture of 1- (6-bromopyridin-3-yl) piperazine (8a) (320mg,1.33mmol), oxetan-3-one (384mg,5.32mmol) and AcOH (240mg,3.99mmol) in DCE (6mL) was stirred at 55 ℃ for 2.5 h. Then NaBH (OAc) is added3(564mg, 2.66mmol) and the mixture was heated to 65 ℃ and stirred for 2 h. By H2The mixture was diluted with O and extracted with DCM. The extract was washed with brine, Na2SO4Dried and concentrated. The residue was purified by column chromatography on silica gel eluting with DCM/MeOH (100: 1-50: 1) to give the title compound 1- (6-bromopyridin-3-yl) -4- (oxetan-3-yl) piperazine (8 b). MS-ESI (M/z):298,300[ M + 1]]+
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (4- (oxa) hexane Cyclobutan-3-yl) piperazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (8)
The title compound 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (5- (4- (oxetan-3-yl) piperazin-1-yl) pyridin-2-yl) pyrazolo [1,5-a]Pyrimidine (8) was synthesized according to 5, replacing 1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine (5d) with 1- (6-bromopyridin-3-yl) -4- (oxetan-3-yl) piperazine (8 b). MS-ESI (M/z):530[ M +1 [)]+
Example 9
(R) -4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazoles And [1,5-a ]]Pyrimidin-3-yl) pyridin-2-yl) -2-methylmorpholine (9)
Figure BDA0003541883110000481
5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (6-fluoropyridin-3-yl) pyrazolo[1,5-a]Pyrimidine (9a)
Reacting 5- ((1R,5S) -1- (2, 5-di)Fluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3-iodopyrazolo [1,5-a]Pyrimidine (5b) (2.00g,4.57mmol), (6-fluoropyridin-3-yl) boronic acid (1.20g,9.13mmol), PdCl2(dppf)2(334mg,0.456mmol) and K3PO4(2M,18.2mmol) in a mixture of 1, 4-dioxane (90mL) was stirred at 80 ℃ for 1.5 h. The mixture was cooled to room temperature and washed with H2Dilute O and extract with EtOAc. Water and brine wash the extract, Na2SO4Drying and concentrating. The residue was purified by PTLC eluting with DCM/MeOH (15:1) to give the title compound 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]]Hexane-2-yl) -3- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyrimidine (9 a). MS-ESI (M/z) 408[ M + 1]]+
(R) -4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazoles And [1,5-a ]]Pyrimidin-3-yl) pyridin-2-yl) -2-methylmorpholine (9)
Mixing 5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0]Hexane-2-yl) -3- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyrimidine (9a) (12.0mg,0.03mmol), (R) -2-methylmorpholine hydrochloride (41mg,0.3mmol) and K2CO3(82mg,0.6mmol) in a mixture of DMSO (0.5mL) was stirred at 150 ℃ overnight. The mixture is cooled to room temperature H2Diluted O and extracted with EtOAc (2X 5 mL). Water and brine wash the extract, Na2SO4Drying and concentrating. The residue was purified by PTLC eluting with DCM/MeOH (20:1) to give the title compound (R) -4- (5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-2-yl) -2-methylmorpholine (9). MS-ESI (M/z):489[ M + 1]]+
Example 10
4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- 6a]Pyrimidin-3-yl) pyridin-3-yl) -1-imino-1 lambda-thiomorpholine 1-oxide (10)
Figure BDA0003541883110000491
6Benzyl 1- (p-toluenesulfonylimino) -1 λ -thiomorpholine-4-carboxylate 1-oxide (10a)
The title compound, benzyl 1- (p-toluenesulfonylimino) -1. lambda6The 1-oxide of-thiomorpholine-4-carboxylate (10a) is prepared according to the method of the literature Tetrahedron 2014,70, 6613-Buchner 6622. MS-ESI (M/z):423[ M + 1]]+
6Benzyl 1-imino-1. lambda. -thiomorpholine-4-carboxylate 1-oxide (10b)
To benzyl 1- (p-toluenesulfonylimino) -1 lambda6To a solution of (10a) (422mg,1.00mmol) of (E) -thiomorpholine-4-carboxylate 1-oxide in DME (10mL) was added naphthalen-1-yl sodium salt (1.2M,3.2mL) and stirred at-60 ℃ for 10 min. The mixture was diluted with methanol and concentrated. The residue was purified by column chromatography on silica eluting with DCM/MeOH (100: 1-40: 1) to give the title compound benzyl 1-imino-1. lambda6Thiomorpholine-4-carboxylate 1-oxide (10b), MS-ESI (M/z):269[ M +1]+.
6Benzyl 1- ((tert-butoxycarbonyl) imino) -1 λ -thiomorpholine-4-carboxylate 1-oxide (10c)
To benzyl 1-imino-1 lambda at 0 DEG C6-thiomorpholine-4-carboxylate 1-oxide (10b) (500mg,1.86mmol) and (Boc)2To a solution of O in DMF (10mL) was added NaH (60%, 223mg,5.58mmol) slowly and stirred at room temperature for 2 h. The mixture is brought to 0 ℃ with H2Quenching by O and extracting by EA. Washing the extract with brine, Na2SO4Drying and concentrating. The residue was purified by column chromatography on silica gel eluting with PE/EA (10:1 to 4:1) to give the title compound benzyl 1- ((tert-butoxycarbonyl) imino) -1. lambda6-thiomorpholine-4-carboxylate 1-oxide (10 c). MS-ESI (M/z):369[ M + 1]]+
6Tert-butyl (1-oxo-1. lambda. -thiomorpholine-1-methylene) carbamate (10d)
Under hydrogen atmosphere, 1- ((tert-butyl)Oxycarbonyl) imino) -1 lambda6A mixture of-thiomorpholine-4-carboxylate 1-oxide (10C) (570mg) and Pd/C (10%) in THF (10mL) was stirred at room temperature for 3 h. Filtration and concentration gave the title compound tert-butyl (1-oxo-1. lambda6-thiomorpholine-1-methylene) carbamate (10 d). MS-ESI (M/z):235[ M + 1]]+
6Tert-butyl (4- (6-bromopyridin-3-yl) -1-oxo-1. lambda. -thiomorpholine-1-methylene) carbamate (10e)
The title compound tert-butyl (4- (6-bromopyridin-3-yl) -1-oxo-1. lambda6-Thiomorpholine-1-methylene) carbamate (10e) replacement of tert-butylpiperazine-1-carboxylate by tert-butyl (1-oxo-1. lambda. according to the method of synthesis of 6a6-thiomorpholine-1-methylene) carbamate (10 d). MS-ESI (M/z):574[ M +1 [)]+
Tert-butyl (4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazoles 6And [1,5-a ]]Pyrimidin-3-yl) pyridin-3-yl) -1-oxo-1 lambda-thiomorpholin-1-ylidene) carbamate (10f)
The title compound tert-butyl (4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) -1-oxo-1 lambda6-Thiomorpholine-1-methylene) carbamate (10f) was synthesized according to the method of 5 substituting 1- ((6-bromopyridin-3-yl) methyl) -4-methylpiperazine (5d) with tert-butyl (4- (6-bromopyridin-3-yl) -1-oxo-1. lambda6-thiomorpholine-1-methylene) carbamate (10 e). MS-ESI (M/z):622[ M +1 [)]+
4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5- 6a]Pyrimidin-3-yl) pyridin-3-yl) -1-imino-1 lambda-thiomorpholine 1-oxide (10)
The title compound 4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) -1-imino-1 lambda6The thiomorpholine 1-oxide (10) isAccording to the synthesis of 6c, tert-butyl 4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) piperazine-1-carboxylate (6b) was replaced with tert-butyl (4- (6- (5- ((5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [ 3.1.0)]Hexane-2-yl) pyrazolo [1,5-a]Pyrimidin-3-yl) pyridin-3-yl) -1-oxo-1 lambda6-thiomorpholine-1-methylene) carbamate (10 f). MS-ESI (M/z):522[ M +1]+
Examples 11-199 listed in Table 1 are essentially the same as examples 1-10, using either commercially available starting materials or prepared according to literature procedures. The names and structures of examples 11-199 are given in Table 1.
TABLE 1
Figure BDA0003541883110000501
Figure BDA0003541883110000511
Figure BDA0003541883110000521
Figure BDA0003541883110000531
Figure BDA0003541883110000541
Figure BDA0003541883110000551
Figure BDA0003541883110000561
Figure BDA0003541883110000571
Figure BDA0003541883110000581
Figure BDA0003541883110000591
Figure BDA0003541883110000601
Figure BDA0003541883110000611
Figure BDA0003541883110000621
Figure BDA0003541883110000631
Figure BDA0003541883110000641
Figure BDA0003541883110000651
Figure BDA0003541883110000661
Figure BDA0003541883110000671
Figure BDA0003541883110000681
Figure BDA0003541883110000691
Figure BDA0003541883110000701
Biological activity
MTS assay kits were purchased from Promega (Madison, Wis., USA). RPMI-1640 medium, fetal bovine serum and penicillin-streptomycin were purchased from Gibco (San Francisco, California, USA). Dimethyl sulfoxide (DMSO) and puromycin were purchased from Sigma (st. Murine interleukin 3(IL-3) was purchased from CST (Boston, Massachusetts, USA).
The inhibitory effect of the compound on TRK was examined by measuring the inhibitory effect of the compound on the proliferation of KM12 cells. KM12 cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum. The cell fullness reaches 40% -80%, digesting the cells, inoculating the cells into a 96-well plate according to the cell concentration of 1000/well, 37 ℃, 5% CO2And (5) incubating for 24 h. The compounds were added to 96-well plates at different concentrations (final concentrations 10000, 3333.3, 1111.1, 370.4, 123.5, 41.2, 13.7, 4.6 and 1.5nM) at 37 ℃ with 5% CO2Incubate for 72 h. MTS was added to each well at a concentration of 20. mu.L MTS per 100. mu.L of the medium. After 2h incubation, the reaction was stopped by adding 25. mu.L of 10% SDS to each well. The absorbance at 490nm and 650nm was measured with a microplate reader. IC calculation Using GraphPad Prism 5.050
The inhibition effect of the compound on TRK fusion or mutation is detected by measuring the inhibition effect of the compound on the proliferation of Ba/F3 mouse pro-B lymphocytes which are over-expressed by TRK wild-type or mutant fusion proteins (TPM3-TRKA, TPM3-TRKA-G595R, AFAP1-TRKB, ETV6-TRKC and ETV 6-TRKC-G623R). The Ba/F3 engineered strain cells were cultured in 2ug/mL puromycin and 10% fetal calf serumRPMI-1640 medium. The cell fullness reaches 40% -80%, the cells are collected and are respectively processed according to the 3x104/mL(Ba/F3-TPM3-TRKA)、1x105/mL(Ba/F3-TPM3-TRKA-G595R)、1x105/mL(Ba/F3-AFAP1-TRKB)、3x104/mL(Ba/F3-ETV6-TRKC)、3x104The cells were seeded in 96-well plates at a concentration of Ba/F3-ETV6-TRKC-G623R, respectively, at 37 ℃ and 5% CO2Incubate for 4 h. The compounds were added to 96-well plates at different concentrations (final concentrations 10000, 3333.3, 1111.1, 370.4, 123.5, 41.2, 13.7, 4.6 and 1.5nM) at 37 ℃ in 5% CO2Incubate for 72 h. MTS was added to each well at a concentration of 20. mu.L MTS per 100. mu.L of the medium. After 2h incubation, the reaction was stopped by adding 25 μ L of 10% SDS per well. The absorbance at 490nm and 650nm was measured with a microplate reader. IC calculation Using GraphPad Prism 5.0 software50
Selected compounds prepared as described above were tested according to the biological methods described herein. The results are shown in table 2:
TABLE 2
Figure BDA0003541883110000711
Figure BDA0003541883110000721
Figure BDA0003541883110000731
Figure BDA0003541883110000741

Claims (23)

1. A compound of formula (I):
Figure FDA0003541883100000011
or a pharmaceutically acceptable salt thereof, wherein:
when X is N and Y is C, as shown in formula (Ia);
Figure FDA0003541883100000012
when X is C, Y is N, as shown in formula (Ib);
Figure FDA0003541883100000013
R1selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
each R2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclic radical-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA1RB1、-ORA1-C(O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(O)ORA1、-OC(O)RA1、-C(O)NRA1RB1、-NRA1C(O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(O)NRA1RB1、-NRA1C(O)ORB1、-NRA1C(O)NRA1RB1、-NRA1C(S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(O)rRA1、-S(O)(=NRE1)RB1、-N=S(O)RA1RB1、-S(O)2ORA1、-OS(O)2RA1、-NRA1S(O)rRB1、-NRA1S(O)(=NRE1)RB1、-S(O)rNRA1RB1、-S(O)(=NRE1)NRA1RB1、-NRA1S(O)2NRA1RB1、-NRA1S(O)(=NRE1)NRA1RB1、-P(O)RA1RB1and-P (O) (OR)A1)(ORB1) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
R3selected from aryl and heteroaryl, wherein each aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from RXSubstituted with the substituent(s);
R4selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA2RB2、-ORA2、-C(O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(O)ORA2、-OC(O)RA2、-C(O)NRA2RB2、-NRA2C(O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(O)NRA2RB2、-NRA2C(O)ORB2、-NRA2C(O)NRA2RB2、-NRA2C(S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(O)rRA2、-S(O)(=NRE2)RB2、-N=S(O)RA2RB2、-S(O)2ORA2、-OS(O)2RA2、-NRA2S(O)rRB2、-NRA2S(O)(=NRE2)RB2、-S(O)rNRA2RB2、-S(O)(=NRE2)NRA2RB2、-NRA2S(O)2NRA2RB2、-NRA2S(O)(=NRE2)NRA2RB2、-P(O)RA2RB2and-P (O) (OR)A2)(ORB2) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or independently selected by at least one RXSubstituted with the substituent(s);
each RA1、RA2、RB1And RB2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RXSubstituted with the substituent(s);
or each "RA1And RB1"or" RA2And RB2Taken together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may be unsubstituted or substituted with 1,2 or 3 heteroatoms selected from RXSubstituted with the substituent(s);
each RE1And RE2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、ORa1、SRa1、-S(O)rRa1、-C(O)Ra1、C(O)ORa1、-C(O)NRa1Rb1and-S (O)rNRa1Rb1Wherein each isThe alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are unsubstituted or substituted with at least one group independently selected from RYSubstituted with the substituent(s);
each RXIndependently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, -NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=N-ORb1)Ra1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tOC(O)Rb1、-(CRc1Rd1)tC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(O)rRb1、-(CRc1Rd1)tS(O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(O)Ra1Rb1、-(CRc1Rd1)tS(O)2ORb1、-(CRc1Rd1)tOS(O)2Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)Rb1、-(CRc1Rd1)tS(O)rNRa1Rb1、-(CRc1Rd1)tS(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)2NRa1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tP(O)Ra1Rb1And- (CR)c1Rd1)tP(O)(ORa1)(ORb1) Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or independently selected by at least one RYSubstituted with the substituent(s);
each Ra1And Rb1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or independently selected from R by at least oneYSubstituted with the substituent(s);
or Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Rc1And Rd1Independent of each otherSelected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one R independently selected from RYSubstituted with the substituent(s);
or Rc1And Rd1Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Re1Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, CN, NO2、-ORa2、-SRa2、-S(O)rRa2、-C(O)Ra2、-C(O)ORa2、-S(O)rNRa2Rb2and-C (O) NRa2Rb2
Each RYIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tC(=NRe2)Ra2、-(CRc2Rd2)tC(=N-ORb2)Ra2、-(CRc2Rd2)tC(O)ORb2、-(CRc2Rd2)tOC(O)Rb2、-(CRc2Rd2)tC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)Rb2、-(CRc2Rd2)tC(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)Rb2、-(CRc2Rd2)tOC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)ORb2、-(CRc2Rd2)tNRa2C(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(S)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)NRa2Rb2、-(CRc2Rd2)tS(O)rRb2、-(CRc2Rd2)tS(O)(=NRe2)Rb2、-(CRc2Rd2)tN=S(O)Ra2Rb2、-(CRc2Rd2)tS(O)2ORb2、-(CRc2Rd2)tOS(O)2Rb2、-(CRc2Rd2)tNRa2S(O)rRb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)Rb2、-(CRc2Rd2)tS(O)rNRa2Rb2、-(CRc2Rd2)tS(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2S(O)2NRa2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tP(O)Ra2Rb2And- (CR)c2Rd2)tP(O)(ORa2)(ORb2) Wherein each of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl isThe radicals being unsubstituted or substituted by at least one radical independently selected from the group consisting of hydroxy, CN, amino, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Ra2And Rb2Independently selected from hydrogen, deuterium, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Ra2And Rb2Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Rc2And Rd2Independently selected from hydrogen, deuterium, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one group independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Rc2And Rd2Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be substituted by 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Re2Independently selected from hydrogen, deuterium, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, -C (O) C1-4Alkyl, -C (O) C3-10Cycloalkyl, -C (O) OC1-4Alkyl, -C (O) OC3-10Cycloalkyl, -C (O) N (C)1-4Alkyl radical)2、-C(O)N(C3-10Cycloalkyl radicals2、-S(O)2C1-4Alkyl, -S (O)2C3-10Cycloalkyl, -S (O)2N(C1-4Alkyl radical)2and-S (O)2N(C3-10Cycloalkyl radicals2
n is selected from 0, 1,2,3 and 4;
each r is independently selected from 0, 1 and 2;
each t is independently selected from 0, 1,2,3, and 4;
the compounds provided do not include:
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-oxadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-oxadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropyl-1, 3, 4-thiadiazole,
2- (tert-butyl) -5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hexan-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) isoindolin-1-one,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1,3, 4-thiadiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5- (trifluoromethyl) -1,3, 4-thiadiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2-isopropylthiazole,
2-cyclopropyl-5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole,
5- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2- (trifluoromethyl) thiazole,
2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -5-isopropylthiazole, and
5-cyclopropyl-2- (5- ((1R,5S) -1- (2, 5-difluorophenyl) -2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) thiazole.
2. The compound of claim 1, wherein X is N, Y is C, the compound is represented by (Ia),
Figure FDA0003541883100000051
wherein R is1、R2、R3、R4And n is as defined for formula (I).
3. The compound of claim 1, wherein X is C, Y is N, and the compound is represented by (Ib),
Figure FDA0003541883100000052
wherein R is1、R2、R3、R4And n is as defined for formula (I).
4. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R3Selected from 5-membered rings.
5. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R3Selected from 6 membered rings.
6. The compound of claim 4, or a pharmaceutically acceptable salt thereof, wherein R3Is selected from
Figure FDA0003541883100000061
Figure FDA0003541883100000062
Which is unsubstituted or substituted by at least one member independently selected from RXIs substituted.
7. The compound of claim 5, or a pharmaceutically acceptable salt thereof, wherein R3Is selected from
Figure FDA0003541883100000063
Figure FDA0003541883100000064
Which is unsubstituted or substituted by at least one member independently selected from RXIs substituted with the substituent(s).
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein R3Substituent R ofXSelected from halogen, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl, heterocyclyl-C1-4Alkyl, - (CR)c1Rd1)tORb1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(O)Ra1And- (CR)c1Rd1)tN=S(O)Ra1Rb1Wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from RYIs substituted.
9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein RYIs selected from C1-10Alkyl radical, C3-10Cycloalkyl, heterocyclyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tS(O)rRb2And- (CR)c2Rd2)tORb2Wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one group independently selected from OH, CN, amino, halogen, C1-10Alkyl and C1-10Substituent of alkoxy.
10. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein R3Substituent R ofXIs selected from-NH2、-CN、-CO2ET, methyl, isopropyl, cyclopropyl,
Figure FDA0003541883100000065
Figure FDA0003541883100000066
Figure FDA0003541883100000071
11. A compound according to any one of claims 1 to 10Or a pharmaceutically acceptable salt thereof, wherein R1Is aryl, wherein aryl is unsubstituted or substituted with at least one substituent independently selected from RXIs substituted with the substituent(s).
12. The compound of claim 11, or a pharmaceutically acceptable salt thereof, wherein R1Is phenyl, wherein phenyl is substituted by halogen.
13. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt thereof, wherein each R2Is independently selected from C1-10Alkyl radical, C3-10Cycloalkyl and halogen, wherein each alkyl and cycloalkyl is unsubstituted or substituted with at least one group independently selected from RXIs substituted with the substituent(s).
14. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt thereof, wherein n is selected from 0 and 1.
15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein R4Selected from hydrogen, halogen, -CN, -NRA2RB2and-C (O) NRA2RB2
16. The compound of claim 15, or a pharmaceutically acceptable salt thereof, wherein R4Selected from hydrogen, Br, -NH2-CN and-C (O) NH2
17. The compound of claim 1 selected from:
Figure FDA0003541883100000072
Figure FDA0003541883100000081
Figure FDA0003541883100000091
Figure FDA0003541883100000101
and pharmaceutically acceptable salts thereof.
18. The compound of claim 1 selected from:
Figure FDA0003541883100000111
Figure FDA0003541883100000121
Figure FDA0003541883100000131
Figure FDA0003541883100000141
and pharmaceutically acceptable salts thereof.
19. A pharmaceutical composition comprising a compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
20. A method of treating, ameliorating, or preventing a condition responsive to inhibition of TRK comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, or at least one pharmaceutical composition thereof, and optionally in combination with a second therapeutic agent.
21. Use of a compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a TRK-mediated disorder.
22. The use of claim 21, wherein the condition includes, but is not limited to, an autoimmune disease, a transplant disease, pain, an infectious disease, or a cell proliferative disease.
23. The use of claim 22, wherein the condition includes, but is not limited to, brain, lung, squamous cell, bladder, stomach, pancreas, breast, head, neck, renal region, kidney, ovary, prostate, colorectal, epidermal, esophageal, testicular, gynecological or thyroid cancer, benign hyperplasia of skin, restenosis and benign prostatic hypertrophy, pancreatitis, kidney disease, chronic and/or acute pain; preventing blastocyst implantation, psoriasis, rashes and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, atherosclerosis, inhibition of keratinocytes responsive to a mixture of growth factors, Chronic Obstructive Pulmonary Disease (COPD) and other diseases.
CN202080063781.2A 2019-09-11 2020-09-10 Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors Pending CN114630829A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962898817P 2019-09-11 2019-09-11
US62/898,817 2019-09-11
PCT/CN2020/114472 WO2021047584A1 (en) 2019-09-11 2020-09-10 SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS

Publications (1)

Publication Number Publication Date
CN114630829A true CN114630829A (en) 2022-06-14

Family

ID=74865837

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080063781.2A Pending CN114630829A (en) 2019-09-11 2020-09-10 Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors

Country Status (2)

Country Link
CN (1) CN114630829A (en)
WO (1) WO2021047584A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102056927A (en) * 2008-05-13 2011-05-11 Irm责任有限公司 Fused nitrogen containing heterocycles and compositions thereof as kinase inhibitors
WO2012034095A1 (en) * 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
WO2019174598A1 (en) * 2018-03-14 2019-09-19 Fochon Pharmaceuticals, Ltd. SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS53350B (en) * 2008-09-22 2014-10-31 Array Biopharma, Inc. Substituted imidazo[1,2b] pyridazine compounds as trk kinase inhibitors
US8637516B2 (en) * 2010-09-09 2014-01-28 Irm Llc Compounds and compositions as TRK inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102056927A (en) * 2008-05-13 2011-05-11 Irm责任有限公司 Fused nitrogen containing heterocycles and compositions thereof as kinase inhibitors
WO2012034095A1 (en) * 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
WO2019174598A1 (en) * 2018-03-14 2019-09-19 Fochon Pharmaceuticals, Ltd. SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
CN111936500A (en) * 2018-03-14 2020-11-13 重庆复创医药研究有限公司 Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors

Also Published As

Publication number Publication date
WO2021047584A1 (en) 2021-03-18

Similar Documents

Publication Publication Date Title
CN110177788B (en) Compounds as BCL-2 selective apoptosis inducers
TWI791511B (en) apoptosis inducer
ES2833576T3 (en) Novel glutaminase inhibitors
CN111936500B (en) Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidines and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
CN108602799B (en) Kinase inhibitor
CN111971287B (en) Macrocyclic compounds as TRK kinase inhibitors
CN115843297A (en) Compounds as BCL-2 inhibitors
CN114008042B (en) Substituted pyrrolo [2,3-b ] pyridine and pyrazolo [3,4-b ] pyridine derivatives as protein kinase inhibitors
CN115551856A (en) Compounds as CDK2/4/6 inhibitors
WO2015151490A1 (en) New tricyclic quinone derivative
CN112771047A (en) Substituted imidazo [1,2-a ] pyridine and [1,2,4] triazolo [1,5-a ] pyridine compounds as RET kinase inhibitors
CN115443277A (en) Compounds as kinase inhibitors
CN106573907A (en) Novel quinoline derivatives and their use in neurodegenerative diseases
CN109153686B (en) Protein kinase inhibitor
CN110139857B (en) Sulfoximine, sulfoximinamide, sulfodiimine, and diimine sulfonamide compounds as indoleamine 2,3-dioxygenase inhibitors
CN108341819B (en) Phosphodiesterase inhibitors and uses thereof
CN109476666B (en) Compound as kinase inhibitor
CN114630829A (en) Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
TW202017927A (en) Substituted [1, 2, 4] triazolo [1, 5-a] pyridine compounds as ret kinase inhibitors
RU2781618C2 (en) Substituted (2-azabicyclo[3.1.0]hexane-2-yl)pyrazolo[1.5-a]pyrimidine and imidazo[1.2-b]pyridazine compounds as trk kinase inhibitors
CN112771042A (en) Nalidinone and pyridylpyrimidinone compounds as protein kinase inhibitors
RU2778294C2 (en) Macrocyclic compounds as trk kinase inhibitors
CN117321051A (en) Compounds as PARP inhibitors
CN115605481A (en) Compounds as kinase inhibitors
CN116964038A (en) Small molecule compounds that bind to tau proteins

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20220825

Address after: 401121 building F, No. 2, Yangliu Road, Yubei District, Chongqing

Applicant after: Chongqing Fushang Yuanchuang Pharmaceutical Technology Co.,Ltd.

Address before: 401121 building F, No. 2, Yangliu Road, Yubei District, Chongqing

Applicant before: FOCHON PHARMACEUTICALS, LTD.

TA01 Transfer of patent application right
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination